Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nature ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961294

RESUMEN

Social communication guides decision-making, which is essential for survival. Social transmission of food preference (STFP) is an ecologically relevant memory paradigm in which an animal learns a desirable food odour from another animal in a social context, creating a long-term memory1,2. How food-preference memory is acquired, consolidated and stored is unclear. Here we show that the posteromedial nucleus of the cortical amygdala (COApm) serves as a computational centre in long-term STFP memory consolidation by integrating social and sensory olfactory inputs. Blocking synaptic signalling by the COApm-based circuit selectively abolished STFP memory consolidation without impairing memory acquisition, storage or recall. COApm-mediated STFP memory consolidation depends on synaptic inputs from the accessory olfactory bulb and on synaptic outputs to the anterior olfactory nucleus. STFP memory consolidation requires protein synthesis, suggesting a gene-expression mechanism. Deep single-cell and spatially resolved transcriptomics revealed robust but distinct gene-expression signatures induced by STFP memory formation in the COApm that are consistent with synapse restructuring. Our data thus define a neural circuit for the consolidation of a socially communicated long-term memory, thereby mechanistically distinguishing protein-synthesis-dependent memory consolidation from memory acquisition, storage or retrieval.

2.
Mol Cell ; 82(24): 4627-4646.e14, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36417913

RESUMEN

Cell lineage specification is accomplished by a concerted action of chromatin remodeling and tissue-specific transcription factors. However, the mechanisms that induce and maintain appropriate lineage-specific gene expression remain elusive. Here, we used an unbiased proteomics approach to characterize chromatin regulators that mediate the induction of neuronal cell fate. We found that Tip60 acetyltransferase is essential to establish neuronal cell identity partly via acetylation of the histone variant H2A.Z. Despite its tight correlation with gene expression and active chromatin, loss of H2A.Z acetylation had little effect on chromatin accessibility or transcription. Instead, loss of Tip60 and acetyl-H2A.Z interfered with H3K4me3 deposition and activation of a unique subset of silent, lineage-restricted genes characterized by a bivalent chromatin configuration at their promoters. Altogether, our results illuminate the mechanisms underlying bivalent chromatin activation and reveal that H2A.Z acetylation regulates neuronal fate specification by establishing epigenetic competence for bivalent gene activation and cell lineage transition.


Asunto(s)
Cromatina , Histonas , Histonas/genética , Histonas/metabolismo , Acetilación , Activación Transcripcional , Cromatina/genética , Procesamiento Proteico-Postraduccional , Nucleosomas
3.
Cell ; 155(3): 621-35, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24243019

RESUMEN

Direct lineage reprogramming is a promising approach for human disease modeling and regenerative medicine, with poorly understood mechanisms. Here, we reveal a hierarchical mechanism in the direct conversion of fibroblasts into induced neuronal (iN) cells mediated by the transcription factors Ascl1, Brn2, and Myt1l. Ascl1 acts as an "on-target" pioneer factor by immediately occupying most cognate genomic sites in fibroblasts. In contrast, Brn2 and Myt1l do not access fibroblast chromatin productively on their own; instead, Ascl1 recruits Brn2 to Ascl1 sites genome wide. A unique trivalent chromatin signature in the host cells predicts the permissiveness for Ascl1 pioneering activity among different cell types. Finally, we identified Zfp238 as a key Ascl1 target gene that can partially substitute for Ascl1 during iN cell reprogramming. Thus, a precise match between pioneer factors and the chromatin context at key target genes is determinative for transdifferentiation to neurons and likely other cell types.


Asunto(s)
Reprogramación Celular , Embrión de Mamíferos/citología , Fibroblastos/citología , Redes Reguladoras de Genes , Neuronas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Factores del Dominio POU/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
4.
Nature ; 521(7552): 352-6, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25830878

RESUMEN

In the context of most induced pluripotent stem (iPS) cell reprogramming methods, heterogeneous populations of non-productive and staggered productive intermediates arise at different reprogramming time points. Despite recent reports claiming substantially increased reprogramming efficiencies using genetically modified donor cells, prospectively isolating distinct reprogramming intermediates remains an important goal to decipher reprogramming mechanisms. Previous attempts to identify surface markers of intermediate cell populations were based on the assumption that, during reprogramming, cells progressively lose donor cell identity and gradually acquire iPS cell properties. Here we report that iPS cell and epithelial markers, such as SSEA1 and EpCAM, respectively, are not predictive of reprogramming during early phases. Instead, in a systematic functional surface marker screen, we find that early reprogramming-prone cells express a unique set of surface markers, including CD73, CD49d and CD200, that are absent in both fibroblasts and iPS cells. Single-cell mass cytometry and prospective isolation show that these distinct intermediates are transient and bridge the gap between donor cell silencing and pluripotency marker acquisition during the early, presumably stochastic, reprogramming phase. Expression profiling reveals early upregulation of the transcriptional regulators Nr0b1 and Etv5 in this reprogramming state, preceding activation of key pluripotency regulators such as Rex1 (also known as Zfp42), Dppa2, Nanog and Sox2. Both factors are required for the generation of the early intermediate state and fully reprogrammed iPS cells, and thus represent some of the earliest known regulators of iPS cell induction. Our study deconvolutes the first steps in a hierarchical series of events that lead to pluripotency acquisition.


Asunto(s)
Separación Celular , Reprogramación Celular/fisiología , Citometría de Flujo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción/metabolismo , 5'-Nucleotidasa/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Receptor Nuclear Huérfano DAX-1/metabolismo , Proteínas de Unión al ADN/metabolismo , Molécula de Adhesión Celular Epitelial , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Integrina alfa4/metabolismo , Antígeno Lewis X/metabolismo , Ratones , Proteína Homeótica Nanog , Proteínas Nucleares/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Tiempo , Factores de Transcripción/análisis
5.
Nat Methods ; 14(6): 621-628, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28504679

RESUMEN

Approaches to differentiating pluripotent stem cells (PSCs) into neurons currently face two major challenges-(i) generated cells are immature, with limited functional properties; and (ii) cultures exhibit heterogeneous neuronal subtypes and maturation stages. Using lineage-determining transcription factors, we previously developed a single-step method to generate glutamatergic neurons from human PSCs. Here, we show that transient expression of the transcription factors Ascl1 and Dlx2 (AD) induces the generation of exclusively GABAergic neurons from human PSCs with a high degree of synaptic maturation. These AD-induced neuronal (iN) cells represent largely nonoverlapping populations of GABAergic neurons that express various subtype-specific markers. We further used AD-iN cells to establish that human collybistin, the loss of gene function of which causes severe encephalopathy, is required for inhibitory synaptic function. The generation of defined populations of functionally mature human GABAergic neurons represents an important step toward enabling the study of diseases affecting inhibitory synaptic transmission.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/fisiología , Proteínas de Homeodominio/genética , Células Madre Pluripotentes/fisiología , Factores de Transcripción/genética , Animales , Ingeniería Celular , Células Cultivadas , Humanos , Ratones , Células Madre Pluripotentes/citología
6.
Methods Mol Biol ; 2683: 39-51, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37300765

RESUMEN

The ability to differentiate pluripotent stem cells and to generate specific cell types is a long-standing goal of regenerative medicine. This can be accomplished by recreating the developmental trajectories using sequential activation of the corresponding signaling pathways, or more recently-by direct programming of cell identities using lineage-specific transcription factors. Notably, to be functional in cell replacement therapies, generation of complex cell types, such as specialized neuronal sub-types of the brain, requires precise induction of molecular profiles and regional specification of the cells. However, the induction of the correct cellular identity and marker gene expression can be hampered by technical challenges, one of which is the robust co-expression of multiple transcription factors that is often required for correct cell identity specification. Here, we describe in detail a method for co-expression of seven transcription factors required for efficient induction of dopaminergic neurons with midbrain characteristics from human embryonic and induced pluripotent stem cells.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células-Madre Neurales , Células Madre Pluripotentes , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neurogénesis/fisiología , Diferenciación Celular/genética
7.
Sci Rep ; 12(1): 22425, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36575212

RESUMEN

The chromodomain helicase DNA-binding protein CHD8 is the most frequently mutated gene in autism spectrum disorder. Despite its prominent disease involvement, little is known about its molecular function in the human brain. CHD8 is a chromatin regulator which binds to the promoters of actively transcribed genes through genomic targeting mechanisms which have yet to be fully defined. By generating a conditional loss-of-function and an endogenously tagged allele in human pluripotent stem cells, we investigated the molecular function and the interaction of CHD8 with chromatin in human neurons. Chromatin accessibility analysis and transcriptional profiling revealed that CHD8 functions as a transcriptional activator at its target genes in human neurons. Furthermore, we found that CHD8 chromatin targeting is cell context-dependent. In human neurons, CHD8 preferentially binds at ETS motif-enriched promoters. This enrichment is particularly prominent on the promoters of genes whose expression significantly changes upon the loss of CHD8. Indeed, among the ETS transcription factors, we identified ELK1 as being most highly correlated with CHD8 expression in primary human fetal and adult cortical neurons and most highly expressed in our stem cell-derived neurons. Remarkably, ELK1 was necessary to recruit CHD8 specifically to ETS motif-containing sites. These findings imply that ELK1 and CHD8 functionally cooperate to regulate gene expression and chromatin states at MAPK/ERK target genes in human neurons. Our results suggest that the MAPK/ERK/ELK1 axis potentially contributes to the pathogenesis caused by CHD8 mutations in human neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Humanos , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Trastorno del Espectro Autista/genética , Cromatina/genética , Cromatina/metabolismo , Neuronas/metabolismo , Factores de Riesgo , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo
8.
Stem Cell Reports ; 16(7): 1763-1776, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34171286

RESUMEN

The differentiation of pluripotent stem cells can be accomplished by sequential activation of signaling pathways or through transcription factor programming. Multistep differentiation imitates embryonic development to obtain authentic cell types, but it suffers from asynchronous differentiation with variable efficiency. Transcription factor programming induces synchronous and efficient differentiation with higher reproducibility but may not always yield authentic cell types. We systematically explored the generation of dopaminergic induced neuronal cells from mouse and human pluripotent stem cells. We found that the proneural factor Ascl1 in combination with mesencephalic factors Lmx1a and Nurr1 induce peripheral dopaminergic neurons. Co-delivery of additional midbrain transcription factors En1, FoxA2, and Pitx3 resulted in facile and robust generation of functional dopaminergic neurons of midbrain character. Our results suggest that more complex combinations of transcription factors may be needed for proper regional specification of induced neuronal cells generated by direct lineage induction.


Asunto(s)
Técnicas de Cultivo de Célula , Neuronas Dopaminérgicas/citología , Mesencéfalo/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Dopamina/metabolismo , Células Madre Embrionarias/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Ratones , Transducción de Señal , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Proteína Wnt1/metabolismo
9.
Neuron ; 107(1): 144-157.e4, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32369733

RESUMEN

During social transmission of food preference (STFP), the combination of an olfactory sensory input with a social cue induces long-term memory of a food odor. How a social cue produces long-term learning of an olfactory input, however, remains unknown. Here we show that the neurons of the anterior olfactory nucleus (AON), which form abundant synaptic projections onto granule cells in the olfactory bulb (OB), express the synaptogenic molecule C1ql3. Deletion of C1ql3 in the dorsolateral AON impaired synaptic AON→OB connections and abolished acquisition, but not recall, of STFP memory without significantly affecting basal olfaction. Moreover, deletion in granule cells of the OB of Bai3, a postsynaptic GPCR that binds C1ql3, similarly suppressed synaptic transmission at AON→OB projections and abolished acquisition, but not recall, of STFP memory. Thus, synaptic AON→OB connections are selectively required for STFP memory acquisition and are formed by an essential interaction of presynaptic C1ql3 with postsynaptic Bai3.


Asunto(s)
Preferencias Alimentarias/fisiología , Aprendizaje/fisiología , Bulbo Olfatorio/fisiología , Vías Olfatorias/fisiología , Transmisión Sináptica/fisiología , Animales , Señales (Psicología) , Glicoproteínas de Membrana/metabolismo , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Receptores de Complemento/metabolismo
10.
Proteins ; 74(2): 368-77, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18623062

RESUMEN

An enzyme with sarcosine dimethylglycine methyltransferase (SDMT) activity has been identified in the thermophilic eukaryote, Galdieria sulphuraria. The crystal structure of the enzyme, solved to a resolution of 1.95 A, revealed a fold highly similar to that of mycolic acid synthases. The kcat and apparent K(M) values were 64.3 min(-1) and 2.0 mM for sarcosine and 85.6 min(-1) and 2.8 mM for dimethylglycine, respectively. Apparent K(M) values of S-adenosylmethionine were 144 and 150 microM for sarcosine and dimethylglycine, respectively, and the enzyme melting temperature was 61.1 degrees C. Modeling of cofactor binding in the active site based on the structure of methoxy mycolic acid synthase 2 revealed a number of conserved interactions within the active site.


Asunto(s)
Proteínas Algáceas/metabolismo , Metiltransferasas/metabolismo , Rhodophyta/enzimología , Sarcosina/análogos & derivados , Sarcosina/metabolismo , Proteínas Algáceas/química , Proteínas Algáceas/aislamiento & purificación , Cristalografía por Rayos X , Estabilidad de Enzimas , Cinética , Metiltransferasas/química , Metiltransferasas/aislamiento & purificación , Pliegue de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia , Especificidad por Sustrato , Temperatura
11.
Cell Stem Cell ; 25(4): 531-541.e6, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585094

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.


Asunto(s)
Deferiprona/uso terapéutico , Células Madre Pluripotentes Inducidas/fisiología , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Proteína Proteolipídica de la Mielina/metabolismo , Oligodendroglía/fisiología , Enfermedad de Pelizaeus-Merzbacher/terapia , Animales , Diferenciación Celular , Células Cultivadas , Ferroptosis , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Peroxidación de Lípido , Ratones , Ratones Mutantes , Mutación/genética , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/efectos de los fármacos , Oligodendroglía/trasplante , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/patología , Trasplante de Células Madre , Reparación del Gen Blanco
12.
Nat Neurosci ; 21(11): 1515-1519, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30349111

RESUMEN

The formation and retrieval of conditioned fear memories critically depend on the amygdala. Here we identify an inhibitory projection from somatostatin-positive neurons in the central amygdala to parvalbumin-positive neurons in the zona incerta that is required for both recent and remote fear memories. Thus, the amygdala inhibitory input to parvalbumin-positive neurons in the zona incerta, a nucleus not previously implicated in fear memory, is an essential component of the fear memory circuitry.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Memoria/fisiología , Zona Incerta/fisiología , Animales , Extinción Psicológica/fisiología , Recuerdo Mental/fisiología , Ratones , Vías Nerviosas/fisiología , Neuronas/fisiología
13.
Technology (Singap World Sci) ; 5(2): 87-97, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28781993

RESUMEN

Neurocircuits in the human brain govern complex behavior and involve connections from many different neuronal subtypes from different brain regions. Recent advances in stem cell biology have enabled the derivation of patient-specific human neuronal cells of various subtypes for the study of neuronal function and disease pathology. Nevertheless, one persistent challenge using these human-derived neurons is the ability to reconstruct models of human brain circuitry. To overcome this obstacle, we have developed a compartmentalized microfluidic device, which allows for spatial separation of cell bodies of different human-derived neuronal subtypes (excitatory, inhibitory and dopaminergic) but is permissive to the spreading of projecting processes. Induced neurons (iNs) cultured in the device expressed pan-neuronal markers and subtype specific markers. Morphologically, we demonstrate defined synaptic contacts between selected neuronal subtypes by synapsin staining. Functionally, we show that excitatory neuronal stimulation evoked excitatory postsynaptic current responses in the neurons cultured in a separate chamber.

14.
Nat Biotechnol ; 35(5): 444-452, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28398344

RESUMEN

Cell replacement therapies for neurodegenerative disease have focused on transplantation of the cell types affected by the pathological process. Here we describe an alternative strategy for Parkinson's disease in which dopamine neurons are generated by direct conversion of astrocytes. Using three transcription factors, NEUROD1, ASCL1 and LMX1A, and the microRNA miR218, collectively designated NeAL218, we reprogram human astrocytes in vitro, and mouse astrocytes in vivo, into induced dopamine neurons (iDANs). Reprogramming efficiency in vitro is improved by small molecules that promote chromatin remodeling and activate the TGFß, Shh and Wnt signaling pathways. The reprogramming efficiency of human astrocytes reaches up to 16%, resulting in iDANs with appropriate midbrain markers and excitability. In a mouse model of Parkinson's disease, NeAL218 alone reprograms adult striatal astrocytes into iDANs that are excitable and correct some aspects of motor behavior in vivo, including gait impairments. With further optimization, this approach may enable clinical therapies for Parkinson's disease by delivery of genes rather than cells.


Asunto(s)
Astrocitos/trasplante , Técnicas de Reprogramación Celular/métodos , Neuronas Dopaminérgicas/citología , Trastornos del Movimiento/prevención & control , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Animales , Astrocitos/citología , Diferenciación Celular/genética , Células Cultivadas , Humanos , Ratones , Trastornos del Movimiento/etiología , Trastornos del Movimiento/patología , Enfermedad de Parkinson/complicaciones , Resultado del Tratamiento
15.
Cell Stem Cell ; 16(1): 39-50, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25467916

RESUMEN

Mutations in the retinoblastoma tumor suppressor gene Rb are involved in many forms of human cancer. In this study, we investigated the early consequences of inactivating Rb in the context of cellular reprogramming. We found that Rb inactivation promotes the reprogramming of differentiated cells to a pluripotent state. Unexpectedly, this effect is cell cycle independent, and instead reflects direct binding of Rb to pluripotency genes, including Sox2 and Oct4, which leads to a repressed chromatin state. More broadly, this regulation of pluripotency networks and Sox2 in particular is critical for the initiation of tumors upon loss of Rb in mice. These studies therefore identify Rb as a global transcriptional repressor of pluripotency networks, providing a molecular basis for previous reports about its involvement in cell fate pliability, and implicate misregulation of pluripotency factors such as Sox2 in tumorigenesis related to loss of Rb function.


Asunto(s)
Carcinogénesis/patología , Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Proteína de Retinoblastoma/metabolismo , Animales , Carcinogénesis/metabolismo , Ciclo Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Unión Proteica , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/deficiencia , Factores de Transcripción SOXB1/genética
16.
Nat Biotechnol ; 31(5): 434-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23584610

RESUMEN

Transplantation of oligodendrocyte precursor cells (OPCs) is a promising potential therapeutic strategy for diseases affecting myelin. However, the derivation of engraftable OPCs from human pluripotent stem cells has proven difficult and primary OPCs are not readily available. Here we report the generation of induced OPCs (iOPCs) by direct lineage conversion. Forced expression of the three transcription factors Sox10, Olig2 and Zfp536 was sufficient to reprogram mouse and rat fibroblasts into iOPCs with morphologies and gene expression signatures resembling primary OPCs. More importantly, iOPCs gave rise to mature oligodendrocytes that could ensheath multiple host axons when co-cultured with primary dorsal root ganglion cells and formed myelin after transplantation into shiverer mice. We propose direct lineage reprogramming as a viable alternative approach for the generation of OPCs for use in disease modeling and regenerative medicine.


Asunto(s)
Fibroblastos/citología , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Oligodendroglía/fisiología , Células Madre/citología , Células Madre/fisiología , Factores de Transcripción/genética , Animales , Diferenciación Celular , Fibroblastos/fisiología , Mejoramiento Genético/métodos , Ratones , Trasplante de Células Madre/métodos
17.
Cell Stem Cell ; 9(6): 517-25, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-22136927

RESUMEN

Cellular plasticity is a major focus of investigation in developmental biology. The recent discovery that induced neuronal (iN) cells can be generated from mouse and human fibroblasts by expression of defined transcription factors suggested that cell fate plasticity is much wider than previously anticipated. In this review, we summarize the most recent developments in this nascent field and suggest criteria to help define and categorize iN cells that take into account the complexity of neuronal identity.


Asunto(s)
Diferenciación Celular/fisiología , Neuronas/fisiología , Animales , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Neuronas/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
J Biotechnol ; 153(3-4): 77-85, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21458505

RESUMEN

Human embryonic stem cells (hESC) possess great potential for applications in regenerative medicine due to their ability to differentiate into any cell type in the body. However, it is crucial to remove residual undifferentiated hESC from the differentiated population to avoid teratoma formation in vivo. The monoclonal antibody, mAb 84, has been shown to bind and kill undifferentiated hESC and is very useful for the elimination of contaminating undifferentiated hESC prior to transplantation. As mAb 84 is an IgM, its large size may impede penetration into embryoid bodies (EB) or cell clumps. To improve penetration, four antibody fragment formats of mAb 84 were engineered and expressed in Escherichia coli: Fab 84, scFv 84, scFv 84-diabody and scFv 84-HTH. All 4 fragments bound specifically to hESC, but only scFv 84-HTH, a single chain variable fragment with a dimerizing helix-turn-helix motif, could recapitulate the cytotoxicity of mAb 84 on multiple hESC lines. The results suggest that multivalency and flexibility between the antigen-binding sites may be essential features required for killing of hESC by mAb 84 and its derivatives. Imaging of EB treated with scFv 84-HTH or mAb 84 showed an even distribution of scFv 84-HTH throughout the EB whereas mAb 84 was localized more to the periphery.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Separación Celular/métodos , Citotoxinas/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Fragmentos de Inmunoglobulinas/farmacología , Proteínas Recombinantes de Fusión/farmacología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/aislamiento & purificación , Sitios de Unión de Anticuerpos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Línea Celular , Citotoxinas/biosíntesis , Citotoxinas/genética , Citotoxinas/aislamiento & purificación , Células Madre Embrionarias/metabolismo , Escherichia coli/genética , Escherichia coli/inmunología , Citometría de Flujo , Humanos , Fragmentos de Inmunoglobulinas/biosíntesis , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/aislamiento & purificación , Ratones , Unión Proteica , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA