Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 27(2): 108841, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38318374

RESUMEN

Zebrafish regenerate their fins which involves a component of cell plasticity. It is currently unclear how regenerate cells divide labor to allow for appropriate growth and patterning. Here, we studied lineage relationships of fluorescence-activated cell sorting-enriched epidermal, bone-forming (osteoblast), and (non-osteoblast) blastemal fin regenerate cells by single-cell RNA sequencing, lineage tracing, targeted osteoblast ablation, and electron microscopy. Most osteoblasts in the outgrowing regenerate derive from osterix+ osteoblasts, while mmp9+ cells reside at segment joints. Distal blastema cells contribute to distal osteoblast progenitors, suggesting compartmentalization of the regenerating appendage. Ablation of osterix+ osteoblasts impairs segment joint and bone matrix formation and decreases regenerate length which is partially compensated for by distal regenerate cells. Our study characterizes expression patterns and lineage relationships of rare fin regenerate cell populations, indicates inherent detection and compensation of impaired regeneration, suggests variable dependence on growth factor signaling, and demonstrates zonation of the elongating fin regenerate.

2.
Nat Cell Biol ; 26(2): 181-193, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38177284

RESUMEN

Mammalian developmental timing is adjustable in vivo by preserving pre-implantation embryos in a dormant state called diapause. Inhibition of the growth regulator mTOR (mTORi) pauses mouse development in vitro, yet how embryonic dormancy is maintained is not known. Here we show that mouse embryos in diapause are sustained by using lipids as primary energy source. In vitro, supplementation of embryos with the metabolite L-carnitine balances lipid consumption, puts the embryos in deeper dormancy and boosts embryo longevity. We identify FOXO1 as an essential regulator of the energy balance in dormant embryos and propose, through meta-analyses of dormant cell signatures, that it may be a common regulator of dormancy across adult tissues. Our results lift a constraint on in vitro embryo survival and suggest that lipid metabolism may be a critical metabolic transition relevant for longevity and stem cell function across tissues.


Asunto(s)
Embrión de Mamíferos , Metabolismo de los Lípidos , Animales , Ratones , Desarrollo Embrionario/fisiología , Metabolismo Energético , Mamíferos
3.
Cell Syst ; 13(9): 768-779.e4, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36044898

RESUMEN

Biological systems have the capacity to not only build and robustly maintain complex structures but also to rapidly break up and rebuild such structures. Here, using primitive societies of Polistes wasps, we show that both robust specialization and rapid plasticity are emergent properties of multi-scale dynamics. We combine theory with experiments that, after perturbing the social structure by removing the queen, correlate time-resolved multi-omics with video recordings. We show that the queen-worker dimorphism relies on the balance between the development of a molecular queen phenotype in all insects and colony-scale inhibition of this phenotype via asymmetric interactions. This allows Polistes to be stable against intrinsic perturbations of molecular states while reacting plastically to extrinsic cues affecting the whole society. Long-term stability of the social structure is reinforced by dynamic DNA methylation. Our study provides a general principle of how both specialization and plasticity can be achieved in biological systems. A record of this paper's transparent peer review process is included in the supplemental information.


Asunto(s)
Avispas , Animales , Metilación de ADN , Fenotipo , Avispas/genética
4.
Front Cell Dev Biol ; 10: 1054476, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589749

RESUMEN

Lineage tracing experiments give dynamic information on the functional behaviour of dividing cells. These experiments therefore have become an important tool for studying stem and progenitor cell fate behavior in vivo. When cell proliferation is high or the frequency of induced clones cannot be precisely controlled, the merging and fragmentation of clones renders the retrospective interpretation of clonal fate data highly ambiguous, potentially leading to unguarded interpretations about lineage relationships and fate behaviour. Here, we discuss and generalize statistical strategies to detect, resolve and make use of clonal fragmentation and merging. We first explain how to detect the rates of clonal fragmentation and merging using simple statistical estimates. We then discuss ways to restore the clonal provenance of labelled cells algorithmically and statistically and elaborate on how the process of clonal fragmentation can indirectly inform about cell fate. We generalize and extend results from the context of their original publication.

5.
Ann N Y Acad Sci ; 1506(1): 74-97, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34605044

RESUMEN

Single cell biology has the potential to elucidate many critical biological processes and diseases, from development and regeneration to cancer. Single cell analyses are uncovering the molecular diversity of cells, revealing a clearer picture of the variation among and between different cell types. New techniques are beginning to unravel how differences in cell state-transcriptional, epigenetic, and other characteristics-can lead to different cell fates among genetically identical cells, which underlies complex processes such as embryonic development, drug resistance, response to injury, and cellular reprogramming. Single cell technologies also pose significant challenges relating to processing and analyzing vast amounts of data collected. To realize the potential of single cell technologies, new computational approaches are needed. On March 17-19, 2021, experts in single cell biology met virtually for the Keystone eSymposium "Single Cell Biology" to discuss advances both in single cell applications and technologies.


Asunto(s)
Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Congresos como Asunto/tendencias , Desarrollo Embrionario/fisiología , Informe de Investigación , Análisis de la Célula Individual/tendencias , Animales , Linaje de la Célula/fisiología , Humanos , Macrófagos/fisiología , Análisis de la Célula Individual/métodos
6.
Nucleic Acids Res ; 49(5): 2609-2628, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33619545

RESUMEN

In most taxa, halving of chromosome numbers during meiosis requires that homologous chromosomes (homologues) pair and form crossovers. Crossovers emerge from the recombination-mediated repair of programmed DNA double-strand breaks (DSBs). DSBs are generated by SPO11, whose activity requires auxiliary protein complexes, called pre-DSB recombinosomes. To elucidate the spatiotemporal control of the DSB machinery, we focused on an essential SPO11 auxiliary protein, IHO1, which serves as the main anchor for pre-DSB recombinosomes on chromosome cores, called axes. We discovered that DSBs restrict the DSB machinery by at least four distinct pathways in mice. Firstly, by activating the DNA damage response (DDR) kinase ATM, DSBs restrict pre-DSB recombinosome numbers without affecting IHO1. Secondly, in their vicinity, DSBs trigger IHO1 depletion mainly by another DDR kinase, ATR. Thirdly, DSBs enable homologue synapsis, which promotes the depletion of IHO1 and pre-DSB recombinosomes from synapsed axes. Finally, DSBs and three DDR kinases, ATM, ATR and PRKDC, enable stage-specific depletion of IHO1 from all axes. We hypothesize that these four negative feedback pathways protect genome integrity by ensuring that DSBs form without excess, are well-distributed, and are restricted to genomic locations and prophase stages where DSBs are functional for promoting homologue pairing and crossover formation.


Asunto(s)
Roturas del ADN de Doble Cadena , Meiosis/genética , ATPasas Asociadas con Actividades Celulares Diversas/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/fisiología , Emparejamiento Cromosómico , Retroalimentación Fisiológica , Gametogénesis , Ratones , Fase Paquiteno , Cromosomas Sexuales , Transducción de Señal
7.
Nat Rev Genet ; 22(1): 59-66, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33024290

RESUMEN

DNA methylation is a key layer of epigenetic regulation. The deposition of methylation marks relies on the catalytic activity of DNA methyltransferases (DNMTs), and their active removal relies on the activity of ten-eleven translocation (TET) enzymes. Paradoxically, in important biological contexts these antagonistic factors are co-expressed and target overlapping genomic regions. The ensuing cyclic biochemistry of cytosine modifications gives rise to a continuous, out-of-thermal equilibrium transition through different methylation states. But what is the purpose of this intriguing turnover of DNA methylation? Recent evidence demonstrates that methylation turnover is enriched at gene distal regulatory elements, including enhancers, and can give rise to large-scale oscillatory dynamics. We discuss this phenomenon and propose that DNA methylation turnover might facilitate key lineage decisions.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Linaje de la Célula , Citosina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Humanos , Oxigenasas de Función Mixta/genética , Proteínas Proto-Oncogénicas/genética
8.
Sci Adv ; 6(18): eaaz5424, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32426477

RESUMEN

The cellular basis and extent of neural stem cell (NSC) self-renewal in adult vertebrates, and their heterogeneity, remain controversial. To explore the functional behavior and dynamics of individual NSCs, we combined genetic lineage tracing, quantitative clonal analysis, intravital imaging, and global population assessments in the adult zebrafish telencephalon. Our results are compatible with a model where adult neurogenesis is organized in a hierarchy in which a subpopulation of deeply quiescent reservoir NSCs with long-term self-renewal potential generate, through asymmetric divisions, a pool of operational NSCs activating more frequently and taking stochastic fates biased toward neuronal differentiation. Our data further suggest the existence of an additional, upstream, progenitor population that supports the continuous generation of new reservoir NSCs, thus contributing to their overall expansion. Hence, we propose that the dynamics of vertebrate neurogenesis relies on a hierarchical organization where growth, self-renewal, and neurogenic functions are segregated between different NSC types.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Animales , Diferenciación Celular , Neurogénesis , Telencéfalo , Pez Cebra
9.
Development ; 147(1)2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31908317

RESUMEN

Zebrafish display widespread and pronounced adult neurogenesis, which is fundamental for their regeneration capability after central nervous system injury. However, the cellular identity and the biological properties of adult newborn neurons are elusive for most brain areas. Here, we have used short-term lineage tracing of radial glia progeny to prospectively isolate newborn neurons from the her4.1+ radial glia lineage in the homeostatic adult forebrain. Transcriptome analysis of radial glia, newborn neurons and mature neurons using single cell sequencing identified distinct transcriptional profiles, including novel markers for each population. Specifically, we detected two separate newborn neuron types, which showed diversity of cell fate commitment and location. Further analyses showed that these cell types are homologous to neurogenic cells in the mammalian brain, identified neurogenic commitment in proliferating radial glia and indicated that glutamatergic projection neurons are generated in the adult zebrafish telencephalon. Thus, we prospectively isolated adult newborn neurons from the adult zebrafish forebrain, identified markers for newborn and mature neurons in the adult brain, and revealed intrinsic heterogeneity among adult newborn neurons and their homology with mammalian adult neurogenic cell types.


Asunto(s)
Encéfalo/citología , Linaje de la Célula , Células Ependimogliales/citología , Neurogénesis , Neuronas/citología , Pez Cebra/anatomía & histología , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos/anatomía & histología , Diencéfalo/citología , Perfilación de la Expresión Génica , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Telencéfalo/citología , Pez Cebra/crecimiento & desarrollo
10.
Development ; 146(12)2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31142540

RESUMEN

During mouse embryogenesis, progenitors within the liver known as hepatoblasts give rise to adult hepatocytes and cholangiocytes. Hepatoblasts, which are specified at E8.5-E9.0, have been regarded as a homogeneous progenitor population that initiate differentiation from E13.5. Recently, scRNA-seq analysis has identified sub-populations of transcriptionally distinct hepatoblasts at E11.5. Here, we show that hepatoblasts are not only transcriptionally but also functionally heterogeneous, and that a subpopulation of E9.5-E10.0 hepatoblasts exhibit a previously unidentified early commitment to cholangiocyte fate. Importantly, we also identify a subpopulation constituting 2% of E9.5-E10.0 hepatoblasts that express the adult stem cell marker Lgr5, and generate both hepatocyte and cholangiocyte progeny that persist for the lifespan of the mouse. Combining lineage tracing and scRNA-seq, we show that Lgr5 marks E9.5-E10.0 bipotent liver progenitors residing at the apex of a hepatoblast hierarchy. Furthermore, isolated Lgr5+ hepatoblasts can be clonally expanded in vitro into embryonic liver organoids, which can commit to either hepatocyte or cholangiocyte fates. Our study demonstrates functional heterogeneity within E9.5 hepatoblasts and identifies Lgr5 as a marker for a subpopulation of bipotent liver progenitors.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hepatocitos/citología , Hígado/embriología , Receptores Acoplados a Proteínas G/metabolismo , Alelos , Animales , Secuencia de Bases , Recuento de Células , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Desarrollo Embrionario , Células Epiteliales/citología , Femenino , Hepatocitos/metabolismo , Homeostasis , Masculino , Ratones , Microscopía Confocal , Células Madre/citología
11.
Eur J Neurosci ; 48(12): 3597-3605, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30408243

RESUMEN

The optic cup houses multipotent retinal progenitor cells that proliferate and differentiate to form the mature retina, containing five main types of neurons and a single glial cell type, the Müller cell. Progenitors of the zebrafish optic cup generate clones that vary regarding the number and types of neurons, a process we previously showed could be described by stochastic models. Here, we present data indicating that each retinal progenitor cell, in the 24 hrs post-fertilization optic cup, is predestined to form a single Müller cell. This striking fate assignment of Müller cells reveals a dual nature of retinal lineages where stochastic mechanisms produce variable numbers of neurons while there is a strong deterministic component governing the formation of glia cells. A possible mechanism for this stereotypic fate assignment could be the maintenance of a clonal backbone during retina development, which would be similar to invertebrate and rodent cortical neurogenesis.


Asunto(s)
Células Ependimogliales/metabolismo , Neuroglía/metabolismo , Retina/metabolismo , Células Madre/metabolismo , Animales , Animales Modificados Genéticamente/genética , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
12.
Sci Rep ; 8(1): 15374, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337647

RESUMEN

ß-cell replacement has been proposed as an effective treatment for some forms of diabetes, and in vitro methods for ß-cell generation are being extensively explored. A potential source of ß-cells comes from fate conversion of exocrine pancreatic cells into the endocrine lineage, by overexpression of three regulators of pancreatic endocrine formation and ß-cell identity, Ngn3, Pdx1 and MafA. Pancreatic ductal organoid cultures have recently been developed that can be expanded indefinitely, while maintaining the potential to differentiate into the endocrine lineage. Here, using mouse pancreatic ductal organoids, we see that co-expression of Ngn3, Pdx1 and MafA are required and sufficient to generate cells that express insulin and resemble ß-cells transcriptome-wide. Efficiency of ß-like cell generation can be significantly enhanced by preventing phosphorylation of Ngn3 protein and further augmented by conditions promoting differentiation. Taken together, our new findings underline the potential of ductal organoid cultures as a source material for generation of ß-like cells and demonstrate that post-translational regulation of reprogramming factors can be exploited to enhance ß-cell generation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Reprogramación Celular , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Organoides/metabolismo , Conductos Pancreáticos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células HEK293 , Humanos , Células Secretoras de Insulina/citología , Ratones , Proteínas del Tejido Nervioso/genética , Organoides/citología , Conductos Pancreáticos/citología , Fosforilación
13.
Cell Syst ; 7(1): 63-76.e12, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30031774

RESUMEN

Pluripotency is accompanied by the erasure of parental epigenetic memory, with naïve pluripotent cells exhibiting global DNA hypomethylation both in vitro and in vivo. Exit from pluripotency and priming for differentiation into somatic lineages is associated with genome-wide de novo DNA methylation. We show that during this phase, co-expression of enzymes required for DNA methylation turnover, DNMT3s and TETs, promotes cell-to-cell variability in this epigenetic mark. Using a combination of single-cell sequencing and quantitative biophysical modeling, we show that this variability is associated with coherent, genome-scale oscillations in DNA methylation with an amplitude dependent on CpG density. Analysis of parallel single-cell transcriptional and epigenetic profiling provides evidence for oscillatory dynamics both in vitro and in vivo. These observations provide insights into the emergence of epigenetic heterogeneity during early embryo development, indicating that dynamic changes in DNA methylation might influence early cell fate decisions.


Asunto(s)
Metilación de ADN/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular , Reprogramación Celular , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Embrión de Mamíferos/citología , Epigénesis Genética/genética , Epigenómica , Genoma , Impresión Genómica , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología
14.
Dev Cell ; 46(3): 360-375.e5, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30057275

RESUMEN

Pancreas development involves a coordinated process in which an early phase of cell segregation is followed by a longer phase of lineage restriction, expansion, and tissue remodeling. By combining clonal tracing and whole-mount reconstruction with proliferation kinetics and single-cell transcriptional profiling, we define the functional basis of pancreas morphogenesis. We show that the large-scale organization of mouse pancreas can be traced to the activity of self-renewing precursors positioned at the termini of growing ducts, which act collectively to drive serial rounds of stochastic ductal bifurcation balanced by termination. During this phase of branching morphogenesis, multipotent precursors become progressively fate-restricted, giving rise to self-renewing acinar-committed precursors that are conveyed with growing ducts, as well as ductal progenitors that expand the trailing ducts and give rise to delaminating endocrine cells. These findings define quantitatively how the functional behavior and lineage progression of precursor pools determine the large-scale patterning of pancreatic sub-compartments.


Asunto(s)
Linaje de la Célula , Células Endocrinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Organogénesis/fisiología , Páncreas/crecimiento & desarrollo , Células Acinares/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Morfogénesis/fisiología , Células Madre/metabolismo
15.
Nat Phys ; 14(5): 469-474, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29736183

RESUMEN

The emergence of complex organs is driven by the coordinated proliferation, migration and differentiation of precursor cells. The fate behaviour of these cells is reflected in the time evolution their progeny, termed clones, which serve as a key experimental observable. In adult tissues, where cell dynamics is constrained by the condition of homeostasis, clonal tracing studies based on transgenic animal models have advanced our understanding of cell fate behaviour and its dysregulation in disease (1, 2). But what can be learned from clonal dynamics in development, where the spatial cohesiveness of clones is impaired by tissue deformations during tissue growth? Drawing on the results of clonal tracing studies, we show that, despite the complexity of organ development, clonal dynamics may converge to a critical state characterized by universal scaling behaviour of clone sizes. By mapping clonal dynamics onto a generalization of the classical theory of aerosols, we elucidate the origin and range of scaling behaviours and show how the identification of universal scaling dependences may allow lineage-specific information to be distilled from experiments. Our study shows the emergence of core concepts of statistical physics in an unexpected context, identifying cellular systems as a laboratory to study non-equilibrium statistical physics.

16.
Nat Cell Biol ; 20(6): 677-687, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29784917

RESUMEN

Recent lineage tracing studies have revealed that mammary gland homeostasis relies on unipotent stem cells. However, whether and when lineage restriction occurs during embryonic mammary development, and which signals orchestrate cell fate specification, remain unknown. Using a combination of in vivo clonal analysis with whole mount immunofluorescence and mathematical modelling of clonal dynamics, we found that embryonic multipotent mammary cells become lineage-restricted surprisingly early in development, with evidence for unipotency as early as E12.5 and no statistically discernable bipotency after E15.5. To gain insights into the mechanisms governing the switch from multipotency to unipotency, we used gain-of-function Notch1 mice and demonstrated that Notch activation cell autonomously dictates luminal cell fate specification to both embryonic and basally committed mammary cells. These functional studies have important implications for understanding the signals underlying cell plasticity and serve to clarify how reactivation of embryonic programs in adult cells can lead to cancer.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Plasticidad de la Célula , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Receptor Notch1/metabolismo , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Glándulas Mamarias Animales/embriología , Ratones , Ratones Transgénicos , Modelos Genéticos , Morfogénesis , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , Receptor Notch1/genética , Transducción de Señal , Análisis de la Célula Individual , Factores de Tiempo
17.
Cell Stem Cell ; 22(1): 119-127.e3, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29249462

RESUMEN

The clonal complexity of adult stem cell pools is progressively lost during homeostatic turnover in several tissues, suggesting a decrease in the number of stem cells with distinct clonal origins. The functional impact of reduced complexity on stem cell pools, and how different tissue microenvironments may contribute to such a reduction, are poorly understood. Here, we performed clonal multicolor lineage tracing of skeletal muscle stem cells (MuSCs) to address these questions. We found that MuSC clonal complexity is maintained during aging despite heterogenous reductions in proliferative capacity, allowing aged muscle to mount a clonally diverse, albeit diminished, response to injury. In contrast, repeated bouts of tissue repair cause a progressive reduction in MuSC clonal complexity indicative of neutral drift. Consistently, biostatistical modeling suggests that MuSCs undergo symmetric expansions with stochastic fate acquisition during tissue repair. These findings establish distinct principles that underlie stem cell dynamics during homeostatic aging and muscle regeneration.


Asunto(s)
Envejecimiento/fisiología , Homeostasis , Músculo Esquelético/citología , Células Madre/citología , Cicatrización de Heridas , Animales , Adhesión Celular , Linaje de la Célula , Células Clonales , Ratones Endogámicos C57BL , Modelos Biológicos , Regeneración , Procesos Estocásticos
18.
Dev Cell ; 41(3): 274-286.e5, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28457793

RESUMEN

The proneural transcription factor Neurogenin3 (Ngn3) plays a critical role in pancreatic endocrine cell differentiation, although regulation of Ngn3 protein is largely unexplored. Here we demonstrate that Ngn3 protein undergoes cyclin-dependent kinase (Cdk)-mediated phosphorylation on multiple serine-proline sites. Replacing wild-type protein with a phosphomutant form of Ngn3 increases α cell generation, the earliest endocrine cell type to be formed in the developing pancreas. Moreover, un(der)phosphorylated Ngn3 maintains insulin expression in adult ß cells in the presence of elevated c-Myc and enhances endocrine specification during ductal reprogramming. Mechanistically, preventing multi-site phosphorylation enhances both Ngn3 stability and DNA binding, promoting the increased expression of target genes that drive differentiation. Therefore, multi-site phosphorylation of Ngn3 controls its ability to promote pancreatic endocrine differentiation and to maintain ß cell function in the presence of pro-proliferation cues and could be manipulated to promote and maintain endocrine differentiation in vitro and in vivo.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso/metabolismo , Animales , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Páncreas/metabolismo , Fosforilación , Transducción de Señal/fisiología
19.
Nat Commun ; 8: 14684, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248284

RESUMEN

Wound healing is essential to repair the skin after injury. In the epidermis, distinct stem cells (SCs) populations contribute to wound healing. However, how SCs balance proliferation, differentiation and migration to repair a wound remains poorly understood. Here, we show the cellular and molecular mechanisms that regulate wound healing in mouse tail epidermis. Using a combination of proliferation kinetics experiments and molecular profiling, we identify the gene signatures associated with proliferation, differentiation and migration in different regions surrounding the wound. Functional experiments show that SC proliferation, migration and differentiation can be uncoupled during wound healing. Lineage tracing and quantitative clonal analysis reveal that, following wounding, progenitors divide more rapidly, but conserve their homoeostatic mode of division, leading to their rapid depletion, whereas SCs become active, giving rise to new progenitors that expand and repair the wound. These results have important implications for tissue regeneration, acute and chronic wound disorders.


Asunto(s)
Movimiento Celular , Epidermis/patología , Células Madre/citología , Cicatrización de Heridas , Animales , Polaridad Celular , Proliferación Celular , Forma de la Célula , Células Clonales , Folículo Piloso/patología , Ratones , Modelos Biológicos , Células Madre/metabolismo
20.
Curr Opin Cell Biol ; 43: 38-45, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27474807

RESUMEN

The coordination of cell proliferation and differentiation is central to the development and maintenance of tissues, while its dysregulation underlies the transition to diseased states. By combining lineage tracing with transcriptional profiling and marker-based assays, statistical methods are delivering insights into the dynamics of stem cells and their developmental precursors. These studies have provided evidence for molecular heterogeneity and fate priming, and have revealed a role for stochasticity in stem cell fate, refocusing the search for regulatory mechanisms. At the same time, they present a quantitative platform to study the initiation and progression of disease. Here, we review how quantitative lineage tracing strategies are shaping our understanding of the cellular mechanisms of tissue development, maintenance and disease.


Asunto(s)
Células/patología , Enfermedad , Organogénesis , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Humanos , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...