Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
2.
Stem Cell Res Ther ; 14(1): 223, 2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37649117

RESUMEN

BACKGROUND: Induced pluripotent stem cells (iPSC) can be differentiated to cells in all three germ layers, as well as cells in the extraembryonic tissues. Efforts in iPSC differentiation into pancreatic progenitors in vitro have largely been focused on optimizing soluble growth cues in conventional two-dimensional (2D) culture, whereas the impact of three-dimensional (3D) matrix properties on the morphogenesis of iPSC remains elusive. METHODS: In this work, we employ gelatin-based thiol-norbornene photo-click hydrogels for in situ 3D differentiation of human iPSCs into pancreatic progenitors (PP). Molecular analysis and single-cell RNA-sequencing were utilized to elucidate on the distinct identities of subpopulations within the 2D and 3D differentiated cells. RESULTS: We found that, while established soluble cues led to predominately PP cells in 2D culture, differentiation of iPSCs using the same soluble factors led to prominent branching morphogenesis, ductal network formation, and generation of diverse endoderm populations. Through single-cell RNA-sequencing, we found that 3D differentiation resulted in enrichments of pan-endodermal cells and ductal cells. We further noted the emergence of a group of extraembryonic cells in 3D, which was absent in 2D differentiation. The unexpected emergence of extraembryonic cells in 3D was found to be associated with enrichment of Wnt and BMP signaling pathways, which may have contributed to the emergence of diverse cell populations. The expressions of PP signature genes PDX1 and NKX6.1 were restored through inhibition of Wnt signaling at the beginning of the posterior foregut stage. CONCLUSIONS: To our knowledge, this work established the first 3D hydrogel system for in situ differentiation of human iPSCs into PPs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Páncreas , Diferenciación Celular , Hidrogeles , ARN
3.
Cell Stem Cell ; 30(7): 950-961.e7, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37419105

RESUMEN

Mechanosensitive hair cells in the cochlea are responsible for hearing but are vulnerable to damage by genetic mutations and environmental insults. The paucity of human cochlear tissues makes it difficult to study cochlear hair cells. Organoids offer a compelling platform to study scarce tissues in vitro; however, derivation of cochlear cell types has proven non-trivial. Here, using 3D cultures of human pluripotent stem cells, we sought to replicate key differentiation cues of cochlear specification. We found that timed modulations of Sonic Hedgehog and WNT signaling promote ventral gene expression in otic progenitors. Ventralized otic progenitors subsequently give rise to elaborately patterned epithelia containing hair cells with morphology, marker expression, and functional properties consistent with both outer and inner hair cells in the cochlea. These results suggest that early morphogenic cues are sufficient to drive cochlear induction and establish an unprecedented system to model the human auditory organ.


Asunto(s)
Proteínas Hedgehog , Células Madre Pluripotentes , Humanos , Proteínas Hedgehog/metabolismo , Cóclea , Células Ciliadas Auditivas Internas , Organoides , Diferenciación Celular/fisiología
4.
Development ; 150(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37381908

RESUMEN

The inner ear sensory epithelia contain mechanosensitive hair cells and supporting cells. Both cell types arise from SOX2-expressing prosensory cells, but the mechanisms underlying the diversification of these cell lineages remain unclear. To determine the transcriptional trajectory of prosensory cells, we established a SOX2-2A-ntdTomato human embryonic stem cell line using CRISPR/Cas9, and performed single-cell RNA-sequencing analyses with SOX2-positive cells isolated from inner ear organoids at various time points between differentiation days 20 and 60. Our pseudotime analysis suggests that vestibular type II hair cells arise primarily from supporting cells, rather than bi-fated prosensory cells in organoids. Moreover, ion channel- and ion-transporter-related gene sets were enriched in supporting cells versus prosensory cells, whereas Wnt signaling-related gene sets were enriched in hair cells versus supporting cells. These findings provide valuable insights into how prosensory cells give rise to hair cells and supporting cells during human inner ear development, and may provide a clue to promote hair cell regeneration from resident supporting cells in individuals with hearing loss or balance disorders.


Asunto(s)
Células Ciliadas Vestibulares , Vestíbulo del Laberinto , Humanos , Organoides , Células Ciliadas Auditivas , Diferenciación Celular/genética
5.
Res Sq ; 2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37163050

RESUMEN

Background Induced pluripotent stem cells (iPSC) can be differentiated to cells in all three germ layers, as well as cells in the extraembryonic tissues. Efforts in iPSC differentiation into pancreatic progenitors in vitro have largely been focused on optimizing soluble growth cues in conventional two-dimensional (2D) culture, whereas the impact of three-dimensional (3D) matrix properties on the morphogenesis of iPSC remains elusive. Methods In this work, we employ gelatin-based thiol-norbornene photo-click hydrogels for in situ 3D differentiation of human iPSCs into pancreatic progenitors (PP). Molecular analysis and single cell RNA-sequencing were utilized to elucidate on the distinct identities of subpopulations within the 2D and 3D differentiated cells. Results We found that, while established soluble cues led to predominately PP cells in 2D culture, differentiation of iPSCs using the same soluble factors led to prominent branching morphogenesis, ductal network formation, and generation of diverse endoderm populations. Through single-cell RNA-sequencing, we found that 3D differentiation resulted in enrichments of pan-endodermal cells and ductal cells. We further noted the emergence of a group of extraembryonic cells in 3D, which was absent in 2D differentiation. The unexpected emergence of extraembryonic cells in 3D was found to be associated with enrichment of Wnt and BMP signaling pathways, which may have contributed to the emergence of diverse cell populations. The expressions of PP signature genes PDX1 and NKX6.1 were restored through inhibition of Wnt signaling at the beginning of the posterior foregut stage. Conclusions To our knowledge, this work established the first 3D hydrogel system for in situ differentiation of human iPSCs into PPs. Ongoing work focuses on enhancing pancreatic differentiation efficiency through modulating physicochemical properties of the iPSC-laden matrices.

6.
Nat Commun ; 13(1): 7053, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396635

RESUMEN

Mutations in CHD7 cause CHARGE syndrome, affecting multiple organs including the inner ear in humans. We investigate how CHD7 mutations affect inner ear development using human pluripotent stem cell-derived organoids as a model system. We find that loss of CHD7 or its chromatin remodeling activity leads to complete absence of hair cells and supporting cells, which can be explained by dysregulation of key otic development-associated genes in mutant otic progenitors. Further analysis of the mutant otic progenitors suggests that CHD7 can regulate otic genes through a chromatin remodeling-independent mechanism. Results from transcriptome profiling of hair cells reveal disruption of deafness gene expression as a potential underlying mechanism of CHARGE-associated sensorineural hearing loss. Notably, co-differentiating CHD7 knockout and wild-type cells in chimeric organoids partially rescues mutant phenotypes by restoring otherwise severely dysregulated otic genes. Taken together, our results suggest that CHD7 plays a critical role in regulating human otic lineage specification and hair cell differentiation.


Asunto(s)
Síndrome CHARGE , Oído Interno , Humanos , Organoides/metabolismo , Neurogénesis , Síndrome CHARGE/genética , Cabello/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
7.
Hum Genet ; 141(3-4): 347-362, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34342719

RESUMEN

Sensorineural hearing loss (SNHL) is a major cause of functional disability in both the developed and developing world. While hearing aids and cochlear implants provide significant benefit to many with SNHL, neither targets the cellular and molecular dysfunction that ultimately underlies SNHL. The successful development of more targeted approaches, such as growth factor, stem cell, and gene therapies, will require a yet deeper understanding of the underlying molecular mechanisms of human hearing and deafness. Unfortunately, the human inner ear cannot be biopsied without causing significant, irreversible damage to the hearing or balance organ. Thus, much of our current understanding of the cellular and molecular biology of human deafness, and of the human auditory system more broadly, has been inferred from observational and experimental studies in animal models, each of which has its own advantages and limitations. In 2013, researchers described a protocol for the generation of inner ear organoids from pluripotent stem cells (PSCs), which could serve as scalable, high-fidelity alternatives to animal models. Here, we discuss the advantages and limitations of conventional models of the human auditory system, describe the generation and characteristics of PSC-derived inner ear organoids, and discuss several strategies and recent attempts to model hereditary deafness in vitro. Finally, we suggest and discuss several focus areas for the further, intensive characterization of inner ear organoids and discuss the translational applications of these novel models of the human inner ear.


Asunto(s)
Sordera , Oído Interno , Pérdida Auditiva Sensorineural , Sordera/genética , Sordera/patología , Pruebas Auditivas , Humanos , Organoides/patología
8.
Methods Mol Biol ; 2520: 135-150, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34724191

RESUMEN

The sensory epithelia of the inner ear contain mechanosensitive hair cells that detect sound and head acceleration. This protocol details a 3D differentiation method to generate inner ear organoids containing sensory epithelia with hair cells. Human pluripotent stem cells are aggregated in low-binding 96-well plates and treated in chemically defined media with extracellular matrix to promote epithelialization. Small molecules and recombinant proteins are applied in a stepwise manner to recapitulate the morphogenic cues (BMP, TGF-ß, FGF, and WNT) present during inner ear development in vivo. These treatments induce the sequential formation of nonneural ectoderm, otic-epibranchial progenitor domain, and otic placodes. The derived otic placodes then undergo self-guided morphogenesis to form otic vesicles, which eventually give rise to sensory epithelia containing hair cells and supporting cells, as well as neurons with synaptic formations to hair cells. This human stem cell-derived inner ear organoid system provides an ideal platform to study human inner ear development and disease in vitro.


Asunto(s)
Oído Interno , Células Madre Pluripotentes , Diferenciación Celular/fisiología , Oído Interno/metabolismo , Células Ciliadas Auditivas , Humanos , Organoides
9.
Methods Cell Biol ; 159: 303-321, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32586448

RESUMEN

The sensory epithelia of the inner ear contain mechanosensitive hair cells that transmit sound, gravity and head motion signals. This protocol describes an in vitro 3D differentiation method, by which the inner ear sensory epithelium harboring hair cells are derived from human pluripotent stem cells (hPSCs). To begin the differentiation, hPSCs are aggregated in low-binding 96-well plates and treated with extracellular matrix proteins to promote epithelialization. By recapitulating signaling pathway activation and attenuation during in vivo inner ear development, the aggregates are treated with small molecules and recombinant proteins that modulate signaling pathways such as BMP, FGF and WNT in a stepwise manner. These treatments induce sequential formation of non-neural ectoderm (NNE), otic-epibranchial progenitor domain (OEPD), and otic placodes. The otic placodes subsequently undergo self-guided morphogenesis to form otic vesicles, which eventually give rise to sensory epithelia containing inner ear hair cells and supporting cells, as well as neurons forming synapses with the hair cells. These hPSC-derived inner ear sensory structures are designated human inner ear organoids. As human inner ear biopsies are nearly impossible to obtain without causing severe injuries to the auditory system of the patients, the human inner ear organoid system provides a powerful in vitro platform for studying human inner ear disease and development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Oído Interno/citología , Organoides/citología , Células Madre Pluripotentes/citología , Diferenciación Celular , Células Cultivadas , Criopreservación , Humanos , Transducción de Señal
10.
Stem Cell Reports ; 14(6): 996-1008, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32442531

RESUMEN

Sensorineural hearing loss and vestibular dysfunction are caused by damage to neurons and mechanosensitive hair cells, which do not regenerate to any clinically relevant extent in humans. Several protocols have been devised to direct pluripotent stem cells (PSCs) into inner ear hair cells and neurons, which display many properties of their native counterparts. The efficiency, reproducibility, and scalability of these protocols are enhanced by incorporating knowledge of inner ear development. Modeling human diseases in vitro through genetic manipulation of PSCs is already feasible, thereby permitting the elucidation of mechanistic understandings of a wide array of disease etiologies. Early studies on transplantation of PSC-derived otic progenitors have been successful in certain animal models, yet restoration of function and long-term cell survival remain unrealized. Through further research, PSC-based approaches will continue to revolutionize our understanding of inner ear biology and contribute to the development of therapeutic treatments for inner ear disorders.


Asunto(s)
Pérdida Auditiva Sensorineural/terapia , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre/métodos , Animales , Oído Interno/citología , Oído Interno/fisiología , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis , Células Madre Pluripotentes/citología
11.
Stem Cell Reports ; 13(1): 147-162, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31204303

RESUMEN

Mutations in the gene encoding the type II transmembrane protease 3 (TMPRSS3) cause human hearing loss, although the underlying mechanisms that result in TMPRSS3-related hearing loss are still unclear. We combined the use of stem cell-derived inner ear organoids with single-cell RNA sequencing to investigate the role of TMPRSS3. Defective Tmprss3 leads to hair cell apoptosis without altering the development of hair cells and the formation of the mechanotransduction apparatus. Prior to degeneration, Tmprss3-KO hair cells demonstrate reduced numbers of BK channels and lower expressions of genes encoding calcium ion-binding proteins, suggesting a disruption in intracellular homeostasis. A proteolytically active TMPRSS3 was detected on cell membranes in addition to ER of cells in inner ear organoids. Our in vitro model recapitulated salient features of genetically associated inner ear abnormalities and will serve as a powerful tool for studying inner ear disorders.


Asunto(s)
Oído Interno/patología , Células Ciliadas Auditivas Internas/patología , Proteínas de la Membrana/genética , Organoides/citología , Serina Proteasas/genética , Animales , Apoptosis/genética , Membrana Celular/metabolismo , Codón sin Sentido , Técnicas de Inactivación de Genes , Homeostasis/genética , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Mecanotransducción Celular , Proteínas de la Membrana/metabolismo , Ratones , Análisis de Secuencia de ARN , Serina Proteasas/metabolismo , Análisis de la Célula Individual
12.
PLoS One ; 13(1): e0191689, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29370269

RESUMEN

The histone demethylase LSD1 plays a pivotal role in cellular differentiation, particularly in silencing lineage-specific genes. However, little is known about how LSD1 regulates neurosensory differentiation in the inner ear. Here we show that LSD1 interacts directly with the transcription factor Pax2 to form the NuRD co-repressor complex at the Pax2 target gene loci in a mouse otic neuronal progenitor cell line (VOT-N33). VOT-N33 cells expressing a Pax2-response element reporter were GFP-negative when untreated, but became GFP positive after forced differentiation or treatment with a potent LSD inhibitor. Pharmacological inhibition of LSD1 activity resulted in the enrichment of mono- and di-methylation of H3K4, upregulation of sensory neuronal genes and an increase in the number of sensory neurons in mouse inner ear organoids. Together, these results identify the LSD1/NuRD complex as a previously unrecognized modulator for Pax2-mediated neuronal differentiation in the inner ear.


Asunto(s)
Diferenciación Celular/fisiología , Oído Interno/citología , Histona Demetilasas/fisiología , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Células-Madre Neurales/citología , Factor de Transcripción PAX2/metabolismo , Animales , Línea Celular , Oído Interno/metabolismo , Proteínas Fluorescentes Verdes/genética , Histona Demetilasas/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Unión Proteica
13.
Nat Biotechnol ; 35(6): 583-589, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459451

RESUMEN

The derivation of human inner ear tissue from pluripotent stem cells would enable in vitro screening of drug candidates for the treatment of hearing and balance dysfunction and may provide a source of cells for cell-based therapies of the inner ear. Here we report a method for differentiating human pluripotent stem cells to inner ear organoids that harbor functional hair cells. Using a three-dimensional culture system, we modulate TGF, BMP, FGF, and WNT signaling to generate multiple otic-vesicle-like structures from a single stem-cell aggregate. Over 2 months, the vesicles develop into inner ear organoids with sensory epithelia that are innervated by sensory neurons. Additionally, using CRISPR-Cas9, we generate an ATOH1-2A-eGFP cell line to detect hair cell induction and demonstrate that derived hair cells exhibit electrophysiological properties similar to those of native sensory hair cells. Our culture system should facilitate the study of human inner ear development and research on therapies for diseases of the inner ear.


Asunto(s)
Oído Interno/crecimiento & desarrollo , Células Ciliadas Auditivas Internas/fisiología , Organogénesis/fisiología , Organoides/crecimiento & desarrollo , Células Madre Pluripotentes/fisiología , Ingeniería de Tejidos/métodos , Células Cultivadas , Oído Interno/citología , Células Ciliadas Auditivas Internas/citología , Humanos , Organoides/citología
14.
Methods Mol Biol ; 1597: 67-83, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28361311

RESUMEN

The inner ear sensory epithelium harbors mechanosensory hair cells responsible for detecting sound and maintaining balance. This protocol describes a three-dimensional (3D) culture system that efficiently generates inner ear sensory epithelia from aggregates of mouse embryonic stem (mES) cells. By mimicking the activations and repressions of key signaling pathways during in vivo inner ear development, mES cell aggregates are sequentially treated with recombinant proteins and small molecule inhibitors for activating or inhibiting the Bmp, TGFß, Fgf, and Wnt signaling pathways. These stepwise treatments promote mES cells to sequentially differentiate into epithelia representing the non-neural ectoderm, preplacodal ectoderm, otic placodal ectoderm, and ultimately, the hair cell-containing sensory epithelia. The derived hair cells are surrounded by a layer of supporting cells and are innervated by sensory neurons. This in vitro inner ear organoid culture system may serve as a valuable tool in developmental and physiological research, disease modeling, drug testing, and potential cell-based therapies.


Asunto(s)
Diferenciación Celular/fisiología , Oído Interno/citología , Epitelio/fisiología , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas/citología , Células Madre Embrionarias de Ratones/citología , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Ectodermo/citología , Ratones , Organogénesis/fisiología , Organoides/citología , Células Receptoras Sensoriales/citología , Transducción de Señal/fisiología
15.
EMBO Rep ; 18(3): 367-376, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28202491

RESUMEN

Three-dimensional (3D) stem cell differentiation cultures recently emerged as a novel model system for investigating human embryonic development and disease progression in vitro, complementing existing animal and two-dimensional (2D) cell culture models. Organoids, the 3D self-organizing structures derived from pluripotent or somatic stem cells, can recapitulate many aspects of structural organization and functionality of their in vivo organ counterparts, thus holding great promise for biomedical research and translational applications. Importantly, faithful recapitulation of disease and development processes relies on the ability to modify the genomic contents in organoid cells. The revolutionary genome engineering technologies, CRISPR/Cas9 in particular, enable investigators to generate various reporter cell lines for prompt validation of specific cell lineages as well as to introduce disease-associated mutations for disease modeling. In this review, we provide historical overviews, and discuss technical considerations, and potential future applications of genome engineering in 3D organoid models.


Asunto(s)
Ingeniería Genética , Genómica , Organoides/metabolismo , Animales , Diferenciación Celular , Línea Celular , Edición Génica/métodos , Expresión Génica , Genes Reporteros , Ingeniería Genética/métodos , Genómica/métodos , Humanos , Técnicas In Vitro , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Técnicas de Cultivo de Tejidos
16.
PLoS One ; 11(9): e0162508, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27607106

RESUMEN

Stem cell-derived inner ear sensory epithelia are a promising source of tissues for treating patients with hearing loss and dizziness. We recently demonstrated how to generate inner ear sensory epithelia, designated as inner ear organoids, from mouse embryonic stem cells (ESCs) in a self-organizing 3D culture. Here we improve the efficiency of this culture system by elucidating how Wnt signaling activity can drive the induction of otic tissue. We found that a carefully timed treatment with the potent Wnt agonist CHIR99021 promotes induction of otic vesicles-a process that was previously self-organized by unknown mechanisms. The resulting otic-like vesicles have a larger lumen size and contain a greater number of Pax8/Pax2-positive otic progenitor cells than organoids derived without the Wnt agonist. Additionally, these otic-like vesicles give rise to large inner ear organoids with hair cells whose morphological, biochemical and functional properties are indistinguishable from those of vestibular hair cells in the postnatal mouse inner ear. We conclude that Wnt signaling plays a similar role during inner ear organoid formation as it does during inner ear development in the embryo.


Asunto(s)
Oído Interno/metabolismo , Organoides/metabolismo , Técnicas de Cultivo de Tejidos/métodos , Vía de Señalización Wnt , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Agregación Celular/efectos de los fármacos , Oído Interno/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mecanotransducción Celular/efectos de los fármacos , Ratones , Miosina VIIa , Miosinas/metabolismo , Organoides/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Factores de Transcripción SOXB1/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
17.
Nat Commun ; 7: 11508, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27215798

RESUMEN

Inner ear sensory epithelia contain mechanosensitive hair cells that transmit information to the brain through innervation with bipolar neurons. Mammalian hair cells do not regenerate and are limited in number. Here we investigate the potential to generate mechanosensitive hair cells from mouse embryonic stem cells in a three-dimensional (3D) culture system. The system faithfully recapitulates mouse inner ear induction followed by self-guided development into organoids that morphologically resemble inner ear vestibular organs. We find that organoid hair cells acquire mechanosensitivity equivalent to functionally mature hair cells in postnatal mice. The organoid hair cells also progress through a similar dynamic developmental pattern of ion channel expression, reminiscent of two subtypes of native vestibular hair cells. We conclude that our 3D culture system can generate large numbers of fully functional sensory cells which could be used to investigate mechanisms of inner ear development and disease as well as regenerative mechanisms for inner ear repair.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Células Ciliadas Vestibulares/fisiología , Células Madre Embrionarias de Ratones/fisiología , Organoides/fisiología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo/química , Medios de Cultivo/metabolismo , Medios de Cultivo/farmacología , Fenómenos Electrofisiológicos/fisiología , Ratones , Modelos Animales , Organogénesis/fisiología , Organoides/citología , Pirazoles/farmacología , Pirimidinas/farmacología , Proteínas Recombinantes/metabolismo
18.
Methods Mol Biol ; 1341: 391-406, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25822723

RESUMEN

This protocol describes a three-dimensional culture method for generating inner ear sensory epithelia, which comprises sensory hair cells and a concurrently arising neuronal population. Mouse embryonic stem cells are initially plated in 96-well plates with differentiation media; following aggregation, Matrigel is added in order to promote epithelialization. A series of small molecule applications is then used over the first 14 days of culture to guide differentiation towards an otic lineage. After 16-20 days, vesicles containing inner ear sensory hair cells and supporting cells arise from the cultured aggregates. Aggregates may be analyzed using immunohistochemistry and electrophysiology techniques. This system serves as a simple and relatively inexpensive in vitro model of inner ear development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Oído Interno/citología , Células Ciliadas Auditivas/citología , Células Madre Embrionarias de Ratones/citología , Neurogénesis , Organoides/citología , Animales , Ratones
19.
PLoS One ; 10(8): e0135060, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26258652

RESUMEN

Nervous system development relies on the generation of precise numbers of excitatory and inhibitory neurons. The homeodomain transcription factor, T-cell leukemia 3 (Tlx3), functions as the master neuronal fate regulator by instructively promoting the specification of glutamatergic excitatory neurons and suppressing the specification of gamma-aminobutyric acid (GABAergic) neurons. However, how Tlx3 promotes glutamatergic neuronal subtype specification is poorly understood. In this study, we found that Tlx3 directly interacts with the epigenetic co-activator cyclic adenosine monophosphate (cAMP)-response element-binding protein (CREB)-binding protein (CBP) and that the Tlx3 homeodomain is essential for this interaction. The interaction between Tlx3 and CBP was enhanced by the three amino acid loop extension (TALE)-class homeodomain transcription factor, pre-B-cell leukemia transcription factor 3 (Pbx3). Using mouse embryonic stem (ES) cells stably expressing Tlx3, we found that the interaction between Tlx3 and CBP became detectable only after these Tlx3-expressing ES cells were committed to a neural lineage, which coincided with increased Pbx3 expression during neural differentiation from ES cells. Forced expression of mutated Tlx3 lacking the homeodomain in ES cells undergoing neural differentiation resulted in significantly reduced expression of glutamatergic neuronal subtype markers, but had little effect on the expression on pan neural markers. Collectively, our results strongly suggest that functional interplay between Tlx3 and CBP plays a critical role in neuronal subtype specification, providing novel insights into the epigenetic regulatory mechanism that modulates the transcriptional efficacy of a selective set of neuronal subtype-specific genes during differentiation.


Asunto(s)
Proteína de Unión a CREB/genética , Células Madre Embrionarias/metabolismo , Neuronas GABAérgicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Neuronas/metabolismo , Animales , Proteína de Unión a CREB/metabolismo , Diferenciación Celular , Linaje de la Célula/genética , Cromatina/química , Cromatina/metabolismo , Embrión de Mamíferos , Células Madre Embrionarias/citología , Epigénesis Genética , Colorantes Fluorescentes , Fura-2 , Neuronas GABAérgicas/citología , Ácido Glutámico/metabolismo , Células HEK293 , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Neuronas/citología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal
20.
Nat Protoc ; 9(6): 1229-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24784820

RESUMEN

This protocol describes a culture system in which inner-ear sensory tissue is produced from mouse embryonic stem (ES) cells under chemically defined conditions. This model is amenable to basic and translational investigations into inner ear biology and regeneration. In this protocol, mouse ES cells are aggregated in 96-well plates in medium containing extracellular matrix proteins to promote epithelialization. During the first 14 d, a series of precisely timed protein and small-molecule treatments sequentially induce epithelia that represent the mouse embryonic non-neural ectoderm, preplacodal ectoderm and otic vesicle epithelia. Ultimately, these tissues develop into cysts with a pseudostratified epithelium containing inner ear hair cells and supporting cells after 16-20 d. Concurrently, sensory-like neurons generate synapse-like structures with the derived hair cells. We have designated the stem cell-derived epithelia harboring hair cells, supporting cells and sensory-like neurons as inner ear organoids. This method provides a reproducible and scalable means to generate inner ear sensory tissue in vitro.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Oído Interno/crecimiento & desarrollo , Células Madre Embrionarias/citología , Organoides/crecimiento & desarrollo , Animales , Células Madre Embrionarias/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Células Ciliadas Auditivas/citología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...