Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 59(3): 415-430.e8, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38320485

RESUMEN

The early limb bud consists of mesenchymal limb progenitors derived from the lateral plate mesoderm (LPM). The LPM also gives rise to the mesodermal components of the flank and neck. However, the cells at these other levels cannot produce the variety of cell types found in the limb. Taking advantage of a direct reprogramming approach, we find a set of factors (Prdm16, Zbtb16, and Lin28a) normally expressed in the early limb bud and capable of imparting limb progenitor-like properties to mouse non-limb fibroblasts. The reprogrammed cells show similar gene expression profiles and can differentiate into similar cell types as endogenous limb progenitors. The further addition of Lin41 potentiates the proliferation of the reprogrammed cells. These results suggest that these same four factors may play pivotal roles in the specification of endogenous limb progenitors.


Asunto(s)
Extremidades , Proteínas , Ratones , Animales , Proteínas/metabolismo , Fibroblastos , Mesodermo/metabolismo , Esbozos de los Miembros
2.
J Neurosci ; 42(8): 1557-1573, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34965974

RESUMEN

Collagen VI is a key component of muscle basement membranes, and genetic variants can cause monogenic muscular dystrophies. Conversely, human genetic studies recently implicated collagen VI in central nervous system function, with variants causing the movement disorder dystonia. To elucidate the neurophysiological role of collagen VI, we generated mice with a truncation of the dystonia-related collagen α3 VI (COL6A3) C-terminal domain (CTD). These Col6a3CTT mice showed a recessive dystonia-like phenotype in both sexes. We found that COL6A3 interacts with the cannabinoid receptor 1 (CB1R) complex in a CTD-dependent manner. Col6a3CTT mice of both sexes have impaired homeostasis of excitatory input to the basal pontine nuclei (BPN), a motor control hub with dense COL6A3 expression, consistent with deficient endocannabinoid (eCB) signaling. Aberrant synaptic input in the BPN was normalized by a CB1R agonist, and motor performance in Col6a3CTT mice of both sexes was improved by CB1R agonist treatment. Our findings identify a readily therapeutically addressable synaptic mechanism for motor control.SIGNIFICANCE STATEMENT Dystonia is a movement disorder characterized by involuntary movements. We previously identified genetic variants affecting a specific domain of the COL6A3 protein as a cause of dystonia. Here, we created mice lacking the affected domain and observed an analogous movement disorder. Using a protein interaction screen, we found that the affected COL6A3 domain mediates an interaction with the cannabinoid receptor 1 (CB1R). Concordantly, our COL6A3-deficient mice showed a deficit in synaptic plasticity linked to a deficit in cannabinoid signaling. Pharmacological cannabinoid augmentation rescued the motor impairment of the mice. Thus, cannabinoid augmentation could be a promising avenue for treating dystonia, and we have identified a possible molecular mechanism mediating this.


Asunto(s)
Cannabinoides , Colágeno Tipo VI , Distonía , Trastornos Distónicos , Neuronas Motoras , Plasticidad Neuronal , Animales , Cannabinoides/metabolismo , Cannabinoides/farmacología , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Distonía/genética , Distonía/metabolismo , Trastornos Distónicos/genética , Trastornos Distónicos/metabolismo , Femenino , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Mutación , Plasticidad Neuronal/efectos de los fármacos , Receptores de Cannabinoides/genética , Receptores de Cannabinoides/metabolismo
3.
Genesis ; 54(11): 573-581, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27636555

RESUMEN

The FLP/FRT system permits rapid phenotypic screening of homozygous lethal mutations in the context of a viable mosaic fly. Combining this system with ovoD dominant female-sterile transgenes enables efficient production of embryos derived from mutant germline clones lacking maternal contribution from a gene of interest. Two distinct sets of FRT chromosomes, carrying either the mini-white (w + mW.hs ), or rosy (ry+ ) and neomycin (neoR ) transgenes are in common use. Parallel ovoD lines were developed using w + mW.hs FRT insertions on the X and chromosomes 2R and 3L, as well as ry+ , neoR FRT insertions on 2L and 3R. Consequently, mutations isolated on the X, 2R and 3L chromosomes in a ry+ , neoR FRT background, are not amenable to germline clonal analysis without labor-intensive recombination onto chromosome arms containing a w + mW.hs FRT. Here we report the creation of a new ovoD line for the ry+ , neoR FRT insertion at position FRT42D on chromosome 2R, through induced recombination in males. To establish the developmental relevance of this reagent we characterized the maternal-effect phenotypes of novel brother of tout-velu alleles generated on an FRT42D chromosome in a somatic mosaic screen. We find that an apparent null mutation that causes severe defects in somatic tissues has a much milder effect on embryonic patterning, emphasizing the necessity of analyzing mutant phenotypes at multiple developmental stages.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Infertilidad Femenina/genética , N-Acetilglucosaminiltransferasas/genética , Alelos , Animales , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Mutación de Línea Germinal/genética , Humanos , Masculino , Mosaicismo/embriología , Fenotipo , Mutaciones Letales Sintéticas/genética , Transgenes
6.
Nature ; 525(7570): 469-78, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26399828

RESUMEN

Stem cells self-renew and generate specialized progeny through differentiation, but vary in the range of cells and tissues they generate, a property called developmental potency. Pluripotent stem cells produce all cells of an organism, while multipotent or unipotent stem cells regenerate only specific lineages or tissues. Defining stem-cell potency relies upon functional assays and diagnostic transcriptional, epigenetic and metabolic states. Here we describe functional and molecular hallmarks of pluripotent stem cells, propose a checklist for their evaluation, and illustrate how forensic genomics can validate their provenance.


Asunto(s)
Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genómica , Humanos
8.
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
9.
Cell Stem Cell ; 16(3): 323-37, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25748935

RESUMEN

To analyze cellular reprogramming at the single-cell level, mass cytometry was used to simultaneously measure markers of pluripotency, differentiation, cell-cycle status, and cellular signaling throughout the reprogramming process. Time-resolved progression analysis of the resulting data sets was used to construct a continuous molecular roadmap for three independent reprogramming systems. Although these systems varied substantially in Oct4, Sox2, Klf4, and c-Myc stoichiometry, they presented a common set of reprogramming landmarks. Early in the reprogramming process, Oct4(high)Klf4(high) cells transitioned to a CD73(high)CD104(high)CD54(low) partially reprogrammed state. Ki67(low) cells from this intermediate population reverted to a MEF-like phenotype, but Ki67(high) cells advanced through the M-E-T and then bifurcated into two distinct populations: an ESC-like Nanog(high)Sox2(high)CD54(high) population and a mesendoderm-like Nanog(low)Sox2(low)Lin28(high)CD24(high)PDGFR-α(high) population. The methods developed here for time-resolved, single-cell progression analysis may be used for the study of additional complex and dynamic systems, such as cancer progression and embryonic development.


Asunto(s)
Antígenos de Diferenciación , Técnicas de Reprogramación Celular , Citometría de Imagen/métodos , Células Madre Pluripotentes Inducidas , Factores de Transcripción , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
10.
Cell Stem Cell ; 15(6): 707-19, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25456834

RESUMEN

N6-methyl-adenosine (m(6)A) is the most abundant modification on messenger RNAs and is linked to human diseases, but its functions in mammalian development are poorly understood. Here we reveal the evolutionary conservation and function of m(6)A by mapping the m(6)A methylome in mouse and human embryonic stem cells. Thousands of messenger and long noncoding RNAs show conserved m(6)A modification, including transcripts encoding core pluripotency transcription factors. m(6)A is enriched over 3' untranslated regions at defined sequence motifs and marks unstable transcripts, including transcripts turned over upon differentiation. Genetic inactivation or depletion of mouse and human Mettl3, one of the m(6)A methylases, led to m(6)A erasure on select target genes, prolonged Nanog expression upon differentiation, and impaired ESC exit from self-renewal toward differentiation into several lineages in vitro and in vivo. Thus, m(6)A is a mark of transcriptome flexibility required for stem cells to differentiate to specific lineages.


Asunto(s)
Adenina/análogos & derivados , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/metabolismo , Metiltransferasas/metabolismo , Adenina/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Proliferación Celular/genética , Secuencia Conservada/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Metiltransferasas/genética , Ratones , Ratones SCID , Datos de Secuencia Molecular , Mutación/genética , Proteína Homeótica Nanog , Procesamiento Postranscripcional del ARN/genética , ARN Interferente Pequeño/genética , Transcriptoma
12.
Curr Opin Genet Dev ; 22(5): 517-22, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22868177

RESUMEN

The experimental induction of specific cell fates in related or unrelated lineages has fascinated developmental biologists for decades. The evaluation of altered cell fates in response to ectopic expression during embryonic development has been a standard assay for interrogating gene function. However, until recently examples of cell lineage conversions were limited to closely related and primitive cell types. The induction of pluripotency in fibroblasts prominently highlighted that combinations of transcription factors can be extremely powerful and are much more effective than single genes. On the basis of this conclusion we previously identified transcription factor combinations that directly induce functional neuronal cells from mesodermal and endodermal cells. This work has evoked numerous additional studies demonstrating direct lineage conversion into neural and other lineages. Here, we review the generation of neural progenitor cells from fibroblasts, which is the newest addition to the arena of induced cell types. Surprisingly, two fundamentally different approaches have been taken to induce this cell type, one direct approach and another that involves the intermediate generation of a partially reprogrammed pluripotent state.


Asunto(s)
Fibroblastos/citología , Células-Madre Neurales/citología , Neuronas/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Modelos Animales de Enfermedad , Desarrollo Embrionario , Endodermo/citología , Endodermo/metabolismo , Fibroblastos/metabolismo , Expresión Génica , Humanos , Mesodermo/citología , Células Madre/metabolismo , Factores de Transcripción
13.
Proc Natl Acad Sci U S A ; 109(7): 2527-32, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22308465

RESUMEN

We recently showed that defined sets of transcription factors are sufficient to convert mouse and human fibroblasts directly into cells resembling functional neurons, referred to as "induced neuronal" (iN) cells. For some applications however, it would be desirable to convert fibroblasts into proliferative neural precursor cells (NPCs) instead of neurons. We hypothesized that NPC-like cells may be induced using the same principal approach used for generating iN cells. Toward this goal, we infected mouse embryonic fibroblasts derived from Sox2-EGFP mice with a set of 11 transcription factors highly expressed in NPCs. Twenty-four days after transgene induction, Sox2-EGFP(+) colonies emerged that expressed NPC-specific genes and differentiated into neuronal and astrocytic cells. Using stepwise elimination, we found that Sox2 and FoxG1 are capable of generating clonal self-renewing, bipotent induced NPCs that gave rise to astrocytes and functional neurons. When we added the Pou and Homeobox domain-containing transcription factor Brn2 to Sox2 and FoxG1, we were able to induce tripotent NPCs that could be differentiated not only into neurons and astrocytes but also into oligodendrocytes. The transcription factors FoxG1 and Brn2 alone also were capable of inducing NPC-like cells; however, these cells generated less mature neurons, although they did produce astrocytes and even oligodendrocytes capable of integration into dysmyelinated Shiverer brain. Our data demonstrate that direct lineage reprogramming using target cell-type-specific transcription factors can be used to induce NPC-like cells that potentially could be used for autologous cell transplantation-based therapies in the brain or spinal cord.


Asunto(s)
Fibroblastos/citología , Neuronas/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...