Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38772378

RESUMEN

Cell-based ex vivo gene therapy in solid organs, especially the liver, has proven technically challenging. Here, we report a feasible strategy for the clinical application of hepatocyte therapy. We first generated high-quality autologous hepatocytes through the large-scale expansion of patient-derived hepatocytes. Moreover, the proliferating patient-derived hepatocytes, together with the AAV2.7m8 variant identified through screening, enabled CRISPR-Cas9-mediated targeted integration efficiently, achieving functional correction of pathogenic mutations in FAH or OTC. Importantly, these edited hepatocytes repopulated the injured mouse liver at high repopulation levels and underwent maturation, successfully treating mice with tyrosinemia following transplantation. Our study combines ex vivo large-scale cell expansion and gene editing in patient-derived transplantable hepatocytes, which holds potential for treating human liver diseases.

2.
Cell Prolif ; : e13633, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38528645

RESUMEN

Hair cell (HC) damage is a leading cause of sensorineural hearing loss, and in mammals supporting cells (SCs) are unable to divide and regenerate HCs after birth spontaneously. Procollagen C-endopeptidase enhancer 2 (Pcolce2), which encodes a glycoprotein that acts as a functional procollagen C protease enhancer, was screened as a candidate regulator of SC plasticity in our previous study. In the current study, we used adeno-associated virus (AAV)-ie (a newly developed adeno-associated virus that targets SCs) to overexpress Pcolce2 in SCs. AAV-Pcolce2 facilitated SC re-entry into the cell cycle both in cultured cochlear organoids and in the postnatal cochlea. In the neomycin-damaged model, regenerated HCs were detected after overexpression of Pcolce2, and these were derived from SCs that had re-entered the cell cycle. These findings reveal that Pcolce2 may serve as a therapeutic target for the regeneration of HCs to treat hearing loss.

3.
Cell Prolif ; : e13620, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38400824

RESUMEN

Irreversible damage to hair cells (HCs) in the cochlea leads to hearing loss. Cochlear supporting cells (SCs) in the murine cochlea have the potential to differentiate into HCs. Neuron membrane glycoprotein M6B (Gpm6b) as a four-transmembrane protein is a potential regulator of HC regeneration according to our previous research. In this study, we found that AAV-ie-mediated Gpm6b overexpression promoted SC-derived organoid expansion. Enhanced Gpm6b prevented the normal decrease in SC plasticity as the cochlea develops by supporting cells re-entry cell cycle and facilitating the SC-to-HC transformation. Also, overexpression of Gpm6b in the organ of Corti through the round window membrane injection facilitated the trans-differentiation of Lgr5+ SCs into HCs. In conclusion, our results suggest that Gpm6b overexpression promotes HC regeneration and highlights a promising target for hearing repair using the inner ear stem cells combined with AAV.

4.
Adv Sci (Weinh) ; 11(11): e2306788, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38189623

RESUMEN

Mutations in OTOFERLIN (OTOF) lead to the autosomal recessive deafness 9 (DFNB9). The efficacy of adeno-associated virus (AAV)-mediated OTOF gene replacement therapy is extensively validated in Otof-deficient mice. However, the clinical safety and efficacy of AAV-OTOF is not reported. Here, AAV-OTOF is generated using good manufacturing practice and validated its efficacy and safety in mouse and non-human primates in order to determine the optimal injection dose, volume, and administration route for clinical trials. Subsequently, AAV-OTOF is delivered into one cochlea of a 5-year-old deaf patient and into the bilateral cochleae of an 8-year-old deaf patient with OTOF mutations. Obvious hearing improvement is detected by the auditory brainstem response (ABR) and the pure-tone audiometry (PTA) in these two patients. Hearing in the injected ear of the 5-year-old patient can be restored to the normal range at 1 month after AAV-OTOF injection, while the 8-year-old patient can hear the conversational sounds. Most importantly, the 5-year-old patient can hear and recognize speech only through the AAV-OTOF-injected ear. This study is the first to demonstrate the safety and efficacy of AAV-OTOF in patients, expands and optimizes current OTOF-related gene therapy and provides valuable information for further application of gene therapies for deafness.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Humanos , Animales , Ratones , Dependovirus/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/terapia , Audición , Sordera/genética , Sordera/terapia , Terapia Genética
5.
Mol Ther ; 32(1): 204-217, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37952086

RESUMEN

Inner ear hair cells detect sound vibration through the deflection of mechanosensory stereocilia. Cytoplasmic protein TPRN has been shown to localize at the taper region of the stereocilia, and mutations in TPRN cause hereditary hearing loss through an unknown mechanism. Here, using biochemistry and dual stimulated emission depletion microscopy imaging, we show that the TPRN, together with its binding proteins CLIC5 and PTPRQ, forms concentric rings in the taper region of stereocilia. The disruption of TPRN rings, triggered by the competitive inhibition of the interaction of TPRN and CLIC5 or exogenous TPRN overexpression, leads to stereocilia degeneration and severe hearing loss. Most importantly, restoration of the TPRN rings can rescue the damaged auditory function of Tprn knockout mice by exogenously expressing TPRN at an appropriate level in HCs via promoter recombinant adeno-associated virus (AAV). In summary, our results reveal highly structured TPRN rings near the taper region of stereocilia that are crucial for stereocilia function and hearing. Also, TPRN ring restoration in stereocilia by AAV-Tprn effectively repairs damaged hearing, which lays the foundation for the clinical application of AAV-mediated gene therapy in patients with TPRN mutation.


Asunto(s)
Sordera , Pérdida Auditiva , Animales , Humanos , Ratones , Sordera/genética , Audición/genética , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Ratones Noqueados , Proteínas/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Estereocilios/metabolismo
6.
Adv Sci (Weinh) ; 11(3): e2306201, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38014592

RESUMEN

OTOF mutations are the principal causes of auditory neuropathy. There are reports on Otof-related gene therapy in mice, but there is no preclinical research on the drug evaluations. Here, Anc80L65 and the mouse hair cell-specific Myo15 promoter (mMyo15) are used to selectively and effectively deliver human OTOF to hair cells in mice and nonhuman primates to evaluate the efficacy and safety of OTOF gene therapy drugs. A new dual-AAV-OTOF-hybrid strategy to transfer full-length OTOF is generated, which can stably restore hearing in adult OTOFp.Q939*/Q939* mice with profound deafness, with the longest duration being at least 150 days, and the best therapeutic effect without difference in hearing from wild-type mice. An AAV microinjection method into the cochlea of cynomolgus monkeys without hearing impairment is further established and found the OTOF can be safely and effectively driven by the mMyo15 promoter in hair cells. In addition, the therapeutic dose of AAV drugs has no impact on normal hearing and does not cause significant systemic toxicity both in mouse and nonhuman primates. In summary, this study develops a potential gene therapy strategy for DFNB9 patients in the clinic and provides complete, standardized, and systematic research data for clinical research and application.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Adulto , Humanos , Ratones , Animales , Proteínas de la Membrana/genética , Pérdida Auditiva Sensorineural/genética , Primates
7.
Cell Prolif ; 56(11): e13483, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37084708

RESUMEN

Hair cells (HCs) in mammals cannot spontaneously regenerate after damage. Atoh1 overexpression can promote HC regeneration in the postnatal cochlea, but the regenerated HCs do not possess the structural and functional characteristics of HCs in situ. The stereocilia on the apical surface of HCs are the first-level structure for sound conduction, and regeneration of functional stereocilia is the key basis for the reproduction of functional HCs. Espin, as an actin bundling protein, plays an important role in the development and structural maintenance of the stereocilia. Here, we found that the upregulation of Espin by AAV-ie was able to induced the aggregation of actin fibres in Atoh1-induced HCs in both cochlear organoids and explants. In addition, we found that persistent Atoh1 overexpression resulted in impaired stereocilia in both endogenous and newly formed HCs. In contrast, the forced expression of Espin in endogenous and regenerative HCs was able to eliminate the stereocilia damage caused by persistent Atoh1 overexpression. Our study shows that the enhanced expression of Espin can optimize the developmental process of stereocilia in Atoh1-induced HCs and can attenuate the damage to native HCs induced by Atoh1 overexpression. These results suggest an effective method to induce the maturation of stereocilia in regenerative HCs and pave the way for functional HC regeneration via supporting cell transdifferentiation.


Asunto(s)
Actinas , Estereocilios , Animales , Estereocilios/metabolismo , Actinas/metabolismo , Células Ciliadas Auditivas/metabolismo , Cóclea , Epitelio/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Mamíferos/metabolismo
8.
Cell Mol Life Sci ; 80(4): 86, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36917323

RESUMEN

Mechanosensitive hair cells (HCs) in the cochlear sensory epithelium are critical for sound detection and transduction. Mammalian HCs in the cochlea undergo cytogenesis during embryonic development, and irreversible damage to hair cells postnatally is a major cause of deafness. During the development of the organ of Corti, HCs and supporting cells (SCs) originate from the same precursors. In the neonatal cochlea, damage to HCs activates adjacent SCs to act as HC precursors and to differentiate into new HCs. However, the plasticity of SCs to produce new HCs is gradually lost with cochlear development. Here, we delineate an essential role for the guanine nucleotide exchange factor Net1 in SC trans-differentiation into HCs. Net1 overexpression mediated by AAV-ie in SCs promoted cochlear organoid formation and HC differentiation under two and three-dimensional culture conditions. Also, AAV-Net1 enhanced SC proliferation in Lgr5-EGFPCreERT2 mice and HC generation as indicated by lineage tracing of HCs in the cochleae of Lgr5-EGFPCreERT2/Rosa26-tdTomatoloxp/loxp mice. We further found that the up-regulation of Wnt/ß-catenin and Notch signaling in AAV-Net1-transduced cochleae might be responsible for the SC proliferation and HC differentiation. Also, Net1 overexpression in SCs enhanced SC proliferation and HC regeneration and survival after HC damage by neomycin. Taken together, our study suggests that Net1 might serve as a potential target for HC regeneration and that AAV-mediated gene regulation may be a promising approach in stem cell-based therapy in hearing restoration.


Asunto(s)
Transdiferenciación Celular , Células Ciliadas Auditivas , Animales , Ratones , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Cóclea , Ratones Transgénicos
10.
Signal Transduct Target Ther ; 7(1): 109, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35449181

RESUMEN

The cochlea consists of multiple types of cells, including hair cells, supporting cells and spiral ganglion neurons, and is responsible for converting mechanical forces into electric signals that enable hearing. Genetic and environmental factors can result in dysfunctions of cochlear and auditory systems. In recent years, gene therapy has emerged as a promising treatment in animal deafness models. One major challenge of the gene therapy for deafness is to effectively deliver genes to specific cells of cochleae. Here, we screened and identified an AAV-ie mutant, AAV-ie-K558R, that transduces hair cells and supporting cells in the cochleae of neonatal mice with high efficiency. AAV-ie-K558R is a safe vector with no obvious deficits in the hearing system. We found that AAV-ie-K558R can partially restore the hearing loss in Prestin KO mice and, importantly, deliver Atoh1 into cochlear supporting cells to generate hair cell-like cells. Our results demonstrate the clinical potential of AAV-ie-K558R for treating the hearing loss caused by hair cell death.


Asunto(s)
Sordera , Pérdida Auditiva , Animales , Cóclea/metabolismo , Sordera/metabolismo , Sordera/terapia , Terapia Genética , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Pérdida Auditiva/terapia , Ratones
11.
Nat Commun ; 11(1): 5699, 2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33177502

RESUMEN

G-protein-coupled receptors (GPCRs) play important roles in cellular functions. However, their intracellular organization is largely unknown. Through investigation of the cannabinoid receptor 1 (CB1), we discovered periodically repeating clusters of CB1 hotspots within the axons of neurons. We observed these CB1 hotspots interact with the membrane-associated periodic skeleton (MPS) forming a complex crucial in the regulation of CB1 signaling. Furthermore, we found that CB1 hotspot periodicity increased upon CB1 agonist application, and these activated CB1 displayed less dynamic movement compared to non-activated CB1. Our results suggest that CB1 forms periodic hotspots organized by the MPS as a mechanism to increase signaling efficacy upon activation.


Asunto(s)
Encéfalo/citología , Imagen Molecular/métodos , Neuronas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Animales , Axones/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Femenino , Recuperación de Fluorescencia tras Fotoblanqueo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente/métodos , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/análisis , Receptor Cannabinoide CB1/genética
12.
Mol Ther ; 28(9): 2083-2095, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32526202

RESUMEN

Transcription growth factor ß (TGF-ß) signaling-triggered epithelial-to-mesenchymal transition (EMT) process is associated with tumor stemness, metastasis, and chemotherapy resistance. However, the epigenomic basis for TGF-ß-induced EMT remains largely unknown. Here we reveal that HDAC1-mediated global histone deacetylation and the gain of specific histone H3 lysine 27 acetylation (H3K27ac)-marked enhancers are essential for the TGF-ß-induced EMT process. Enhancers gained upon TGF-ß treatment are linked to gene activation of EMT markers and cancer metastasis. Notably, dynamic enhancer gain or loss mainly occurs within pre-existing topologically associated domains (TADs) in epithelial cells, with minimal three-dimensional (3D) genome architecture reorganization. Through motif enrichment analysis of enhancers that are lost or gained upon TGF-ß stimulation, we identify FOXA2 as a key factor to activate epithelial-specific enhancer activity, and we also find that TEAD4 forms a complex with SMAD2/3 to mediate TGF-ß signaling-triggered mesenchymal enhancer reprogramming. Together, our results implicate that key transcription-factor (TF)-mediated enhancer reprogramming modulates the developmental transition in TGF-ß signaling-associated cancer metastasis.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Elementos de Facilitación Genéticos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Células A549 , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Factor Nuclear 3-beta del Hepatocito/metabolismo , Hepatocitos/metabolismo , Histona Desacetilasa 1/metabolismo , Histonas/metabolismo , Humanos , Ratones , Proteínas Musculares/metabolismo , Metástasis de la Neoplasia , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción de Dominio TEA , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
13.
Am J Transl Res ; 12(1): 130-141, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32051742

RESUMEN

Auditory hair cells are the mechanical sensors of sound waves in the inner ear, and the stereocilia, which are actin-rich protrusions of different heights on the apical surfaces of hair cells, are responsible for the transduction of sound waves into electrical signals. As a crucial actin-binding and bundling protein, espin is able to cross-link actin filaments and is therefore necessary for stereocilia morphogenesis. Using advanced super-resolution stimulated emission depletion microscopy, we imaged espin expression at the sub-diffraction limit along the whole length of the stereocilia in outer hair cells and inner hair cells in order to better understand espin's function in the development of stereocilia.

14.
Nat Commun ; 10(1): 3733, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31427575

RESUMEN

Hearing loss is the most common sensory disorder. While gene therapy has emerged as a promising treatment of inherited diseases like hearing loss, it is dependent on the identification of gene delivery vectors. Adeno-associated virus (AAV) vector-mediated gene therapy has been approved in the US for treating a rare inherited eye disease but no safe and efficient vectors have been identified that can target the diverse types of inner ear cells. Here, we identify an AAV variant, AAV-inner ear (AAV-ie), for gene delivery in mouse inner ear. Our results show that AAV-ie transduces the cochlear supporting cells (SCs) with high efficiency, representing a vast improvement over conventional AAV serotypes. Furthermore, after AAV-ie-mediated transfer of the Atoh1 gene, we find that many SCs trans-differentiated into new HCs. Our results suggest that AAV-ie is a useful tool for the cochlear gene therapy and for investigating the mechanism of HC regeneration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Dependovirus/genética , Terapia Genética/métodos , Células Ciliadas Auditivas Internas/citología , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Animales , Células Cultivadas , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Masculino , Ratones , Ratones Endogámicos C57BL
15.
J Mol Cell Biol ; 10(3): 216-228, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29481611

RESUMEN

Clinical therapies of pluripotent stem cells (PSCs)-based transplantation have been hindered by frequent development of teratomas or tumors in animal models and clinical patients. Therefore, clarifying the mechanism of carcinogenesis in stem cell therapy is of great importance for reducing the risk of tumorigenicity. Here we differentiate Oct4-GFP mouse embryonic stem cells (mESCs) into neural progenitor cells (NPCs) and find that a minority of Oct4+ cells are continuously sustained at Oct4+ state. These cells can be enriched and proliferated in a standard ESC medium. Interestingly, the differentiation potential of these enriched cells is tightly restricted with much higher tumorigenic activity, which are thus defined as differentiation-resistant ESCs (DR-ESCs). Transcriptomic and epigenomic analyses show that DR-ESCs are characterized by primordial germ cell-like gene signatures (Dazl, Rec8, Stra8, Blimp1, etc.) and specific epigenetic patterns distinct from mESCs. Moreover, the DR-ESCs possess germ cell potential to generate Sycp3+ haploid cells and are able to reside in sperm-free spermaduct induced by busulfan. Finally, we find that TGFß signaling is overactivated in DR-ESCs, and inhibition of TGFß signaling eliminates the tumorigenicity of mESC-derived NPCs by inducing the full differentiation of DR-ESCs. These data demonstrate that these TGFß-hyperactivated germ cell-like DR-ESCs are the main contributor for the tumorigenicity of ESCs-derived target cell therapy and that inhibition of TGFß signaling in ESC-derived NPC transplantation could drastically reduce the risk of tumor development.


Asunto(s)
Carcinogénesis/metabolismo , Células Madre Embrionarias de Ratones/citología , Células-Madre Neurales/citología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Carcinogénesis/genética , Diferenciación Celular , Línea Celular , Epigénesis Genética , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Células-Madre Neurales/metabolismo , Transcriptoma
16.
J Biol Chem ; 290(7): 4500-11, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548277

RESUMEN

Mouse pluripotent stem cells (PSCs), such as ES cells and induced PSCs (iPSCs), are an excellent system to investigate the molecular and cellular mechanisms involved in early embryonic development. The signaling pathways orchestrated by leukemia inhibitor factor/STAT3, Wnt/ß-catenin, and FGF/MEK/ERK play key roles in the generation of pluripotency. However, the function of TGF-ß signaling in this process remains elusive. Here we show that inhibiting TGF-ß signaling with its inhibitor SB431542 can substitute for Oct4 during reprogramming. Moreover, inhibiting TGF-ß signaling can sustain the pluripotency of iPSCs and ES cells through modulating FGF/MEK/ERK signaling. Therefore, this study reveals a novel function of TGF-ß signaling inhibition in the generation and maintenance of PSCs.


Asunto(s)
Benzamidas/farmacología , Reprogramación Celular/efectos de los fármacos , Dioxoles/farmacología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
17.
Int J Dev Neurosci ; 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23183026

RESUMEN

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

18.
Proc Natl Acad Sci U S A ; 108(29): 12119-24, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730158

RESUMEN

Proliferation of the neural/neuronal progenitor cells (NPCs) at the ventricular zone of the dorsal spinal cord requires the stimuli of Wnt and bone morphogenic protein (BMP). However, how these two signaling pathways are regulated to initiate differentiation in the NPCs as they enter the intermediate zone is not known. Here, we show that Smad6, a negative regulator of BMP signaling, is expressed in the intermediate zone of the chick dorsal spinal cord. Knockdown experiments show that Smad6 is required for promoting NPCs to exit the cell cycle and differentiate into neurons. Although we find that Smad6 inhibits BMP signaling, as expected, we also find that Smad6 unexpectedly inhibits the Wnt/ß-catenin pathway. The inhibition of the Wnt/ß-catenin pathway by Smad6 is independent of its effect on the BMP pathway. Rather, Smad6 through its N-terminal domain and link region enhances the interaction of C-terminal binding protein with the ß-catenin/T cell factor (TCF) complex and the TCF-binding element to inhibit ß-catenin-mediated transcriptional activation. Our study provides evidence that transition of NPCs from a proliferative state to a differentiating state is controlled by the dual inhibitory role of Smad6 to both BMP and Wnt signaling at the level of transcription.


Asunto(s)
Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Tubo Neural/embriología , Neuronas/citología , Transducción de Señal/fisiología , Proteína smad6/metabolismo , Análisis de Varianza , Animales , Western Blotting , Bromodesoxiuridina , Embrión de Pollo , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Electroporación , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/genética , Inmunoprecipitación , Hibridación in Situ , Luciferasas , Tubo Neural/metabolismo , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Proteína smad6/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
19.
Development ; 137(13): 2095-105, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20504958

RESUMEN

Bone morphogenetic protein (BMP) signaling plays a crucial role in maintaining the pluripotency of mouse embryonic stem cells (ESCs) and has negative effects on ESC neural differentiation. However, it remains unclear when and how BMP signaling executes those different functions during neural commitment. Here, we show that a BMP4-sensitive window exists during ESC neural differentiation. Cells at this specific period correspond to the egg cylinder stage epiblast and can be maintained as ESC-derived epiblast stem cells (ESD-EpiSCs), which have the same characteristics as EpiSCs derived from mouse embryos. We propose that ESC neural differentiation occurs in two stages: first from ESCs to ESD-EpiSCs and then from ESD-EpiSCs to neural precursor cells (NPCs). We further show that BMP4 inhibits the conversion of ESCs into ESD-EpiSCs during the first stage, and suppresses ESD-EpiSC neural commitment and promotes non-neural lineage differentiation during the second stage. Mechanistic studies show that BMP4 inhibits FGF/ERK activity at the first stage but not at the second stage; and IDs, as important downstream genes of BMP signaling, partially substitute for BMP4 functions at both stages. We conclude that BMP signaling has distinct functions during different stages of ESC neural commitment.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Neurogénesis , Animales , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Ratones , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...