Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(19)2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37823232

RESUMEN

Neural crest cells generate numerous derivatives, including pigment cells, and are a model for studying how fate specification from multipotent progenitors is controlled. In mammals, the core gene regulatory network for melanocytes (their only pigment cell type) contains three transcription factors, Sox10, Pax3 and Mitf, with the latter considered a master regulator of melanocyte development. In teleosts, which have three to four pigment cell types (melanophores, iridophores and xanthophores, plus leucophores e.g. in medaka), gene regulatory networks governing fate specification are poorly understood, although Mitf function is considered conserved. Here, we show that the regulatory relationships between Sox10, Pax3 and Mitf are conserved in zebrafish, but the role for Mitf is more complex than previously emphasized, affecting xanthophore development too. Similarly, medaka Mitf is necessary for melanophore, xanthophore and leucophore formation. Furthermore, expression patterns and mutant phenotypes of pax3 and pax7 suggest that Pax3 and Pax7 act sequentially, activating mitf expression. Pax7 modulates Mitf function, driving co-expressing cells to differentiate as xanthophores and leucophores rather than melanophores. We propose that pigment cell fate specification should be considered to result from the combinatorial activity of Mitf with other transcription factors.


Asunto(s)
Oryzias , Pez Cebra , Animales , Redes Reguladoras de Genes , Mamíferos/genética , Melanocitos/metabolismo , Mutación , Cresta Neural/metabolismo , Oryzias/genética , Oryzias/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
Development ; 148(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-35020872

RESUMEN

Neural crest cells are crucial in development, not least because of their remarkable multipotency. Early findings stimulated two hypotheses for how fate specification and commitment from fully multipotent neural crest cells might occur, progressive fate restriction (PFR) and direct fate restriction, differing in whether partially restricted intermediates were involved. Initially hotly debated, they remain unreconciled, although PFR has become favoured. However, testing of a PFR hypothesis of zebrafish pigment cell development refutes this view. We propose a novel 'cyclical fate restriction' hypothesis, based upon a more dynamic view of transcriptional states, reconciling the experimental evidence underpinning the traditional hypotheses.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Cresta Neural/crecimiento & desarrollo , Pez Cebra/crecimiento & desarrollo , Animales , Linaje de la Célula/fisiología , Transición Epitelial-Mesenquimal/genética , Regulación del Desarrollo de la Expresión Génica/genética , Melanocitos/metabolismo , Pigmentación/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
3.
Dev Biol ; 471: 76-88, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33326797

RESUMEN

Our lineage tracing studies using multiple Cre mouse lines showed a concurrent labeling of abundant taste bud cells and the underlying connective tissue with a neural crest (NC) origin, warranting a further examination on the issue of whether there is an NC derivation of taste bud cells. In this study, we mapped NC cell lineages in three different models, Sox10-iCreERT2/tdT mouse, GFP+ neural fold transplantation to GFP- chickens, and Sox10-Cre/GFP-RFP zebrafish model. We found that in mice, Sox10-iCreERT2 specifically labels NC cell lineages with a single dose of tamoxifen at E7.5 and that the labeled cells were widely distributed in the connective tissue of the tongue. No labeled cells were found in taste buds or the surrounding epithelium in the postnatal mice. In the GFP+/GFP- chicken chimera model, GFP+ cells migrated extensively to the cranial region of chicken embryos ipsilateral to the surgery side but were absent in taste buds in the base of oral cavity and palate. In zebrafish, Sox10-Cre/GFP-RFP faithfully labeled known NC-derived tissues but did not label taste buds in lower jaw or the barbel. Our data, together with previous findings in axolotl, indicate that taste buds are not derived from NC cells in rodents, birds, amphibians or teleost fish.


Asunto(s)
Linaje de la Célula , Cresta Neural/embriología , Papilas Gustativas/embriología , Animales , Embrión de Pollo , Pollos , Ratones , Ratones Transgénicos , Cresta Neural/citología , Papilas Gustativas/citología , Pez Cebra
4.
PLoS Genet ; 15(2): e1007941, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811380

RESUMEN

Skin pigment patterns are important, being under strong selection for multiple roles including camouflage and UV protection. Pigment cells underlying these patterns form from adult pigment stem cells (APSCs). In zebrafish, APSCs derive from embryonic neural crest cells, but sit dormant until activated to produce pigment cells during metamorphosis. The APSCs are set-aside in an ErbB signaling dependent manner, but the mechanism maintaining quiescence until metamorphosis remains unknown. Mutants for a pigment pattern gene, parade, exhibit ectopic pigment cells localised to the ventral trunk, but also supernumerary cells restricted to the Ventral Stripe. Contrary to expectations, these melanocytes and iridophores are discrete cells, but closely apposed. We show that parade encodes Endothelin receptor Aa, expressed in the blood vessels, most prominently in the medial blood vessels, consistent with the ventral trunk phenotype. We provide evidence that neuronal fates are not affected in parade mutants, arguing against transdifferentiation of sympathetic neurons to pigment cells. We show that inhibition of BMP signaling prevents specification of sympathetic neurons, indicating conservation of this molecular mechanism with chick and mouse. However, inhibition of sympathetic neuron differentiation does not enhance the parade phenotype. Instead, we pinpoint ventral trunk-restricted proliferation of neural crest cells as an early feature of the parade phenotype. Importantly, using a chemical genetic screen for rescue of the ectopic pigment cell phenotype of parade mutants (whilst leaving the embryonic pattern untouched), we identify ErbB inhibitors as a key hit. The time-window of sensitivity to these inhibitors mirrors precisely the window defined previously as crucial for the setting aside of APSCs in the embryo, strongly implicating adult pigment stem cells as the source of the ectopic pigment cells. We propose that a novel population of APSCs exists in association with medial blood vessels, and that their quiescence is dependent upon Endothelin-dependent factors expressed by the blood vessels.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Receptores ErbB/metabolismo , Pigmentos Biológicos/metabolismo , Receptor de Endotelina A/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Receptores ErbB/antagonistas & inhibidores , Melanocitos/citología , Melanocitos/metabolismo , Melanóforos/citología , Melanóforos/metabolismo , Modelos Biológicos , Mutación , Cresta Neural/citología , Cresta Neural/metabolismo , Fenotipo , Receptor de Endotelina A/genética , Transducción de Señal , Pigmentación de la Piel/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
5.
Dev Growth Differ ; 63(9): 516-522, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34807452

RESUMEN

SoxE-type transcription factors, Sox10 and Sox9, are key regulators of the development of neural crest cells. Sox10 specifies pigment cell, glial, and neuronal lineages, whereas Sox9 is reportedly closely associated with skeletogenic lineages in the head, but its involvement in pigment cell formation has not been investigated genetically. Thus, it is not fully understood whether or how distinctly these genes as well as their paralogs in teleosts are subfunctionalized. We have previously shown using the medaka fish Oryzias latipes that pigment cell formation is severely affected by the loss of sox10a, yet unaffected by the loss of sox10b. Here we aimed to determine whether Sox9 is involved in the specification of pigment cell lineage. The sox9b homozygous mutation did not affect pigment cell formation, despite lethality at the early larval stages. By using sox10a, sox10b, and sox9b mutations, compound mutants were established for the sox9b and sox10 genes and pigment cell phenotypes were analyzed. Simultaneous loss of sox9b and sox10a resulted in the complete absence of melanophores and xanthophores from hatchlings and severely defective iridophore formation, as has been previously shown for sox10a-/- ; sox10b-/- double mutants, indicating that Sox9b as well as Sox10b functions redundantly with Sox10a in pigment cell development. Notably, leucophores were present in sox9b-/- ; sox10a-/- and sox10a-/- ; sox10b-/- double mutants, but their numbers were significantly reduced in the sox9b-/- ; sox10a-/- mutants. These findings highlight that Sox9b is involved in pigment cell formation, and plays a more critical role in leucophore development than Sox10b.


Asunto(s)
Linaje de la Célula , Melanóforos , Oryzias , Factor de Transcripción SOX9 , Animales , Cresta Neural , Oryzias/genética , Oryzias/crecimiento & desarrollo , Factor de Transcripción SOX9/genética
6.
PLoS Genet ; 14(10): e1007402, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30286071

RESUMEN

Multipotent neural crest (NC) progenitors generate an astonishing array of derivatives, including neuronal, skeletal components and pigment cells (chromatophores), but the molecular mechanisms allowing balanced selection of each fate remain unknown. In zebrafish, melanocytes, iridophores and xanthophores, the three chromatophore lineages, are thought to share progenitors and so lend themselves to investigating the complex gene regulatory networks (GRNs) underlying fate segregation of NC progenitors. Although the core GRN governing melanocyte specification has been previously established, those guiding iridophore and xanthophore development remain elusive. Here we focus on the iridophore GRN, where mutant phenotypes identify the transcription factors Sox10, Tfec and Mitfa and the receptor tyrosine kinase, Ltk, as key players. Here we present expression data, as well as loss and gain of function results, guiding the derivation of an initial iridophore specification GRN. Moreover, we use an iterative process of mathematical modelling, supplemented with a Monte Carlo screening algorithm suited to the qualitative nature of the experimental data, to allow for rigorous predictive exploration of the GRN dynamics. Predictions were experimentally evaluated and testable hypotheses were derived to construct an improved version of the GRN, which we showed produced outputs consistent with experimentally observed gene expression dynamics. Our study reveals multiple important regulatory features, notably a sox10-dependent positive feedback loop between tfec and ltk driving iridophore specification; the molecular basis of sox10 maintenance throughout iridophore development; and the cooperation between sox10 and tfec in driving expression of pnp4a, a key differentiation gene. We also assess a candidate repressor of mitfa, a melanocyte-specific target of sox10. Surprisingly, our data challenge the reported role of Foxd3, an established mitfa repressor, in iridophore regulation. Our study builds upon our previous systems biology approach, by incorporating physiologically-relevant parameter values and rigorous evaluation of parameter values within a qualitative data framework, to establish for the first time the core GRN guiding specification of the iridophore lineage.


Asunto(s)
Cromatóforos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Cresta Neural/metabolismo , Biología de Sistemas/métodos , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Linaje de la Célula/genética , Cromatóforos/citología , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Mutación , Cresta Neural/citología , Cresta Neural/embriología , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Células Madre/citología , Células Madre/metabolismo , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
PLoS Genet ; 14(4): e1007260, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29621239

RESUMEN

Mechanisms generating diverse cell types from multipotent progenitors are fundamental for normal development. Pigment cells are derived from multipotent neural crest cells and their diversity in teleosts provides an excellent model for studying mechanisms controlling fate specification of distinct cell types. Zebrafish have three types of pigment cells (melanocytes, iridophores and xanthophores) while medaka have four (three shared with zebrafish, plus leucophores), raising questions about how conserved mechanisms of fate specification of each pigment cell type are in these fish. We have previously shown that the Sry-related transcription factor Sox10 is crucial for fate specification of pigment cells in zebrafish, and that Sox5 promotes xanthophores and represses leucophores in a shared xanthophore/leucophore progenitor in medaka. Employing TILLING, TALEN and CRISPR/Cas9 technologies, we generated medaka and zebrafish sox5 and sox10 mutants and conducted comparative analyses of their compound mutant phenotypes. We show that specification of all pigment cells, except leucophores, is dependent on Sox10. Loss of Sox5 in Sox10-defective fish partially rescued the formation of all pigment cells in zebrafish, and melanocytes and iridophores in medaka, suggesting that Sox5 represses Sox10-dependent formation of these pigment cells, similar to their interaction in mammalian melanocyte specification. In contrast, in medaka, loss of Sox10 acts cooperatively with Sox5, enhancing both xanthophore reduction and leucophore increase in sox5 mutants. Misexpression of Sox5 in the xanthophore/leucophore progenitors increased xanthophores and reduced leucophores in medaka. Thus, the mode of Sox5 function in xanthophore specification differs between medaka (promoting) and zebrafish (repressing), which is also the case in adult fish. Our findings reveal surprising diversity in even the mode of the interactions between Sox5 and Sox10 governing specification of pigment cell types in medaka and zebrafish, and suggest that this is related to the evolution of a fourth pigment cell type.


Asunto(s)
Linaje de la Célula , Melanocitos/metabolismo , Oryzias/genética , Pigmentación/genética , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXE/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Alelos , Animales , Regulación del Desarrollo de la Expresión Génica , Melanocitos/citología , Cresta Neural/metabolismo , Factores de Transcripción SOXD/metabolismo , Factores de Transcripción SOXE/metabolismo , Proteínas de Pez Cebra/metabolismo
8.
Int J Mol Sci ; 22(24)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34948326

RESUMEN

The neural crest shows an astonishing multipotency, generating multiple neural derivatives, but also pigment cells, skeletogenic and other cell types. The question of how this process is controlled has been the subject of an ongoing debate for more than 35 years. Based upon new observations of zebrafish pigment cell development, we have recently proposed a novel, dynamic model that we believe goes some way to resolving the controversy. Here, we will firstly summarize the traditional models and the conflicts between them, before outlining our novel model. We will also examine our recent dynamic modelling studies, looking at how these reveal behaviors compatible with the biology proposed. We will then outline some of the implications of our model, looking at how it might modify our views of the processes of fate specification, differentiation, and commitment.


Asunto(s)
Cresta Neural/fisiología , Neurogénesis/fisiología , Animales , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Pez Cebra/fisiología
9.
Development ; 143(8): 1328-39, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26952986

RESUMEN

Mural cells (MCs) consisting of vascular smooth muscle cells and pericytes cover the endothelial cells (ECs) to regulate vascular stability and homeostasis. Here, we clarified the mechanism by which MCs develop and cover ECs by generating transgenic zebrafish lines that allow live imaging of MCs and by lineage tracing in vivo To cover cranial vessels, MCs derived from either neural crest cells or mesoderm emerged around the preformed EC tubes, proliferated and migrated along EC tubes. During their migration, the MCs moved forward by extending their processes along the inter-EC junctions, suggesting a role for inter-EC junctions as a scaffold for MC migration. In the trunk vasculature, MCs derived from mesoderm covered the ventral side of the dorsal aorta (DA), but not the posterior cardinal vein. Furthermore, the MCs migrating from the DA or emerging around intersegmental vessels (ISVs) preferentially covered arterial ISVs rather than venous ISVs, indicating that MCs mostly cover arteries during vascular development. Thus, live imaging and lineage tracing enabled us to clarify precisely how MCs cover the EC tubes and to identify the origins of MCs.


Asunto(s)
Células Endoteliales/citología , Músculo Liso Vascular/citología , Pericitos/citología , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Microscopía Confocal , Pez Cebra
10.
Bioessays ; 39(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28009049

RESUMEN

Skin pigment pattern formation is a paradigmatic example of pattern formation. In zebrafish, the adult body stripes are generated by coordinated rearrangement of three distinct pigment cell-types, black melanocytes, shiny iridophores and yellow xanthophores. A stem cell origin of melanocytes and iridophores has been proposed although the potency of those stem cells has remained unclear. Xanthophores, however, seemed to originate predominantly from proliferation of embryonic xanthophores. Now, data from Singh et al. shows that all three cell-types derive from shared stem cells, and that these cells generate peripheral neural cell-types too. Furthermore, clonal compositions are best explained by a progressive fate restriction model generating the individual cell-types. The numbers of adult pigment stem cells associated with the dorsal root ganglia remain low, but progenitor numbers increase significantly during larval development up to metamorphosis, likely via production of partially restricted progenitors on the spinal nerves.


Asunto(s)
Cresta Neural , Pez Cebra/embriología , Animales , Metamorfosis Biológica , Pigmentación , Células Madre
11.
Dev Biol ; 418(1): 166-178, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27502435

RESUMEN

Vertebrate SoxE genes (Sox8, 9, and 10) are key regulators of neural crest cell (NCC) development. These genes arose by duplication from a single SoxE gene in the vertebrate ancestor. Although SoxE paralogs are coexpressed early in NCC development, later, Sox9 is restricted to skeletogenic lineages in the head, and Sox10 to non-skeletogenic NCC in the trunk and head. When this subfunctionalization evolved and its possible role in the evolution of the neural crest are unknown. Sea lampreys are basal vertebrates that also possess three SoxE genes, while only a single SoxE is present in the cephalochordate amphioxus. In order to address the functional divergence of SoxE genes, and to determine if differences in their biochemical functions may be linked to changes in neural crest developmental potential, we examined the ability of lamprey and amphioxus SoxE genes to regulate differentiation of NCC derivatives in zebrafish colourless (cls) mutants lacking expression of sox10. Our findings suggest that the proto-vertebrate SoxE gene possessed both melanogenic and neurogenic capabilities prior to SoxE gene duplication. Following the agnathan-gnathostome split, lamprey SoxE1 and SoxE3 largely lost their melanogenic and/or enteric neurogenic properties, while gnathostome SoxE paralogs have retained functional conservation. We posit that this difference in protein subfunctionalization is a direct consequence of the independent regulation of SoxE paralog expression between the two lineages. Specifically, we propose that the overlapping expression of gnathostome SoxE paralogs in early neural crest largely constrained the function of gnathostome SoxE proteins. In contrast, the largely non-overlapping expression of lamprey SoxE paralogs allowed them to specialize with regard to their DNA-binding and/or protein interaction properties. Restriction of developmental potential among cranial and trunk neural crest in lampreys may be related to constraints on SoxE activity among duplicates, but such specialization does not appear to have occurred in gnathostomes. This highlights an important difference in the evolution of SoxE activity between these two divergent vertebrate lineages and provides insights for understanding how cell fate restriction in different NCC populations may be dependent on subfunctionalization among SoxE duplicates.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Lampreas/embriología , Anfioxos/embriología , Cresta Neural/embriología , Factores de Transcripción SOXE/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Evolución Biológica , Diferenciación Celular/genética , Duplicación de Gen/genética , Factor de Transcripción Asociado a Microftalmía/biosíntesis , Cresta Neural/citología , Neurogénesis/genética , Proteínas de Pez Cebra/biosíntesis
12.
PLoS Genet ; 10(4): e1004246, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699463

RESUMEN

Mechanisms generating diverse cell types from multipotent progenitors are crucial for normal development. Neural crest cells (NCCs) are multipotent stem cells that give rise to numerous cell-types, including pigment cells. Medaka has four types of NCC-derived pigment cells (xanthophores, leucophores, melanophores and iridophores), making medaka pigment cell development an excellent model for studying the mechanisms controlling specification of distinct cell types from a multipotent progenitor. Medaka many leucophores-3 (ml-3) mutant embryos exhibit a unique phenotype characterized by excessive formation of leucophores and absence of xanthophores. We show that ml-3 encodes sox5, which is expressed in premigratory NCCs and differentiating xanthophores. Cell transplantation studies reveal a cell-autonomous role of sox5 in the xanthophore lineage. pax7a is expressed in NCCs and required for both xanthophore and leucophore lineages; we demonstrate that Sox5 functions downstream of Pax7a. We propose a model in which multipotent NCCs first give rise to pax7a-positive partially fate-restricted intermediate progenitors for xanthophores and leucophores; some of these progenitors then express sox5, and as a result of Sox5 action develop into xanthophores. Our results provide the first demonstration that Sox5 can function as a molecular switch driving specification of a specific cell-fate (xanthophore) from a partially-restricted, but still multipotent, progenitor (the shared xanthophore-leucophore progenitor).


Asunto(s)
Cresta Neural/crecimiento & desarrollo , Oryzias/crecimiento & desarrollo , Pigmentación/genética , Factores de Transcripción SOXD/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica/genética , Melanóforos/fisiología , Cresta Neural/fisiología , Oryzias/fisiología , Factor de Transcripción PAX7/genética , Fenotipo , Pigmentación/fisiología , Células Madre/fisiología
13.
Am J Hum Genet ; 92(3): 415-21, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23395477

RESUMEN

Autosomal-recessive albinism is a hypopigmentation disorder with a broad phenotypic range. A substantial fraction of individuals with albinism remain genetically unresolved, and it has been hypothesized that more genes are to be identified. By using homozygosity mapping of an inbred Faroese family, we identified a 3.5 Mb homozygous region (10q22.2-q22.3) on chromosome 10. The region contains five protein-coding genes, and sequencing of one of these, C10orf11, revealed a nonsense mutation that segregated with the disease and showed a recessive inheritance pattern. Investigation of additional albinism-affected individuals from the Faroe Islands revealed that five out of eight unrelated affected persons had the nonsense mutation in C10orf11. Screening of a cohort of autosomal-recessive-albinism-affected individuals residing in Denmark showed a homozygous 1 bp duplication in C10orf11 in an individual originating from Lithuania. Immunohistochemistry showed localization of C10orf11 in melanoblasts and melanocytes in human fetal tissue, but no localization was seen in retinal pigment epithelial cells. Knockdown of the zebrafish (Danio rerio) homolog with the use of morpholinos resulted in substantially decreased pigmentation and a reduction of the apparent number of pigmented melanocytes. The morphant phenotype was rescued by wild-type C10orf11, but not by mutant C10orf11. In conclusion, we have identified a melanocyte-differentiation gene, C10orf11, which when mutated causes autosomal-recessive albinism in humans.


Asunto(s)
Albinismo/genética , Diferenciación Celular/genética , Cromosomas Humanos Par 10 , Codón sin Sentido , Genes Recesivos , Melanocitos/metabolismo , Albinismo/metabolismo , Albinismo/patología , Animales , Aberraciones Cromosómicas , Mapeo Cromosómico/métodos , Femenino , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Masculino , Melanocitos/patología , Pigmentación/genética , Epitelio Pigmentado de la Retina/metabolismo , Pez Cebra
14.
Development ; 138(16): 3579-89, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21771814

RESUMEN

Coordination of cell proliferation and differentiation is crucial for tissue formation, repair and regeneration. Some tissues, such as skin and blood, depend on differentiation of a pluripotent stem cell population, whereas others depend on the division of differentiated cells. In development and in the hair follicle, pigmented melanocytes are derived from undifferentiated precursor cells or stem cells. However, differentiated melanocytes may also have proliferative capacity in animals, and the potential for differentiated melanocyte cell division in development and regeneration remains largely unexplored. Here, we use time-lapse imaging of the developing zebrafish to show that while most melanocytes arise from undifferentiated precursor cells, an unexpected subpopulation of differentiated melanocytes arises by cell division. Depletion of the overall melanocyte population triggers a regeneration phase in which differentiated melanocyte division is significantly enhanced, particularly in young differentiated melanocytes. Additionally, we find reduced levels of Mitf activity using an mitfa temperature-sensitive line results in a dramatic increase in differentiated melanocyte cell division. This supports models that in addition to promoting differentiation, Mitf also promotes withdrawal from the cell cycle. We suggest differentiated cell division is relevant to melanoma progression because the human melanoma mutation MITF(4T)(Δ)(2B) promotes increased and serial differentiated melanocyte division in zebrafish. These results reveal a novel pathway of differentiated melanocyte division in vivo, and that Mitf activity is essential for maintaining cell cycle arrest in differentiated melanocytes.


Asunto(s)
División Celular , Melanocitos/citología , Melanocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mutación , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Factor de Transcripción Asociado a Microftalmía/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
15.
PLoS Genet ; 7(9): e1002265, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21909283

RESUMEN

The mechanisms generating stably differentiated cell-types from multipotent precursors are key to understanding normal development and have implications for treatment of cancer and the therapeutic use of stem cells. Pigment cells are a major derivative of neural crest stem cells and a key model cell-type for our understanding of the genetics of cell differentiation. Several factors driving melanocyte fate specification have been identified, including the transcription factor and master regulator of melanocyte development, Mitf, and Wnt signalling and the multipotency and fate specification factor, Sox10, which drive mitf expression. While these factors together drive multipotent neural crest cells to become specified melanoblasts, the mechanisms stabilising melanocyte differentiation remain unclear. Furthermore, there is controversy over whether Sox10 has an ongoing role in melanocyte differentiation. Here we use zebrafish to explore in vivo the gene regulatory network (GRN) underlying melanocyte specification and differentiation. We use an iterative process of mathematical modelling and experimental observation to explore methodically the core melanocyte GRN we have defined. We show that Sox10 is not required for ongoing differentiation and expression is downregulated in differentiating cells, in response to Mitfa and Hdac1. Unexpectedly, we find that Sox10 represses Mitf-dependent expression of melanocyte differentiation genes. Our systems biology approach allowed us to predict two novel features of the melanocyte GRN, which we then validate experimentally. Specifically, we show that maintenance of mitfa expression is Mitfa-dependent, and identify Sox9b as providing an Mitfa-independent input to melanocyte differentiation. Our data supports our previous suggestion that Sox10 only functions transiently in regulation of mitfa and cannot be responsible for long-term maintenance of mitfa expression; indeed, Sox10 is likely to slow melanocyte differentiation in the zebrafish embryo. More generally, this novel approach to understanding melanocyte differentiation provides a basis for systematic modelling of differentiation in this and other cell-types.


Asunto(s)
Redes Reguladoras de Genes/genética , Histona Desacetilasa 1/genética , Melanocitos/citología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXE/genética , Proteínas de Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/genética , Animales , Diferenciación Celular , Histona Desacetilasa 1/metabolismo , Melanocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Modelos Teóricos , Cresta Neural/citología , Cresta Neural/crecimiento & desarrollo , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXE/metabolismo , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt/genética , Proteínas de Pez Cebra/metabolismo
16.
J R Soc Interface ; 21(212): 20230537, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38503342

RESUMEN

The challenge to understand differentiation and cell lineages in development has resulted in many bioinformatics software tools, notably those working with gene expression data obtained via single-cell RNA sequencing obtained at snapshots in time. Reconstruction methods for trajectories often proceed by dimension reduction, data clustering and then computation of a tree graph in which edges indicate closely related clusters. Cell lineages can then be deduced by following paths through the tree. In the case of multi-potent cells undergoing differentiation, this trajectory reconstruction involves the reconstruction of multiple distinct lineages corresponding to commitment to each of a set of distinct fates. Recent work suggests that there may be cases in which the cell differentiation process involves trajectories that explore, in a dynamic and oscillatory fashion, propensity to differentiate into a number of possible cell fates before commitment finally occurs. Here, we show theoretically that the presence of such oscillations provides intrinsic constraints on the quality and resolution of the trajectory reconstruction process, even for idealized noise-free data. These constraints point to inherent common limitations of current methodologies and serve both to provide additional challenge in the development of software tools and also may help to understand features observed in recent experiments.


Asunto(s)
Algoritmos , Programas Informáticos , Diferenciación Celular , Biología Computacional/métodos , Análisis de la Célula Individual/métodos
17.
Development ; 137(3): 389-94, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20081187

RESUMEN

Although the regulation of osteoblast and adipocyte differentiation from mesenchymal stem cells has been studied for some time, very little is known about what regulates their appearance in discrete regions of the embryo. Here we show that, as in other vertebrates, zebrafish osteoblasts and adipocytes originate in part from cephalic neural crest (CNC) precursors. We investigated the roles that the retinoic acid (RA) and Peroxisome proliferator-activated receptor gamma (Pparg) pathways play in vivo and found that both pathways act on CNC to direct adipocyte differentiation at the expense of osteoblast formation. In addition, we identify two distinct roles for RA in the osteoblast lineage: an early role in blocking the recruitment of osteoblasts and a later role in mature osteoblasts to promote bone matrix synthesis. These findings might help to increase our understanding of skeletal and obesity-related diseases and aid in the development of stem cell-based regenerative therapies.


Asunto(s)
Cresta Neural/citología , PPAR gamma/fisiología , Tretinoina/fisiología , Adipocitos/citología , Animales , Matriz Ósea/crecimiento & desarrollo , Diferenciación Celular , Linaje de la Célula , Osteoblastos/citología , Células Madre/citología , Pez Cebra
18.
Artículo en Inglés | MEDLINE | ID: mdl-38010612

RESUMEN

It has been 30 (!!) years since I began working on zebrafish pigment cells, as a postdoc in the laboratory of Prof. Christiane Nüsslein-Volhard. There, I participated in the first large-scale mutagenesis screen in zebrafish, focusing on pigment cell mutant phenotypes. The isolation of colourless, shady, parade and choker mutants allowed us (as a postdoc in Prof. Judith Eisen's laboratory, and then in my own laboratory at the University of Bath since 1997) to pursue my ambition to address long-standing problems in the neural crest field. Thus, we have studied how neural crest cells choose individual fates, resulting in our recent proposal of a new, and potentially unifying, model which we call Cyclical Fate Restriction, as well as addressing how pigment cell patterns are generated. A key feature of our work in the last 10 years has been the use of mathematical modelling approaches to clarify our biological models and to refine our interpretations. None of this would have been possible without a hugely talented group of laboratory members and other collaborators from around the world-it has been, and I am sure will continue to be, a pleasure and privilege to work with you all!

19.
Dis Model Mech ; 16(2)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36847161

RESUMEN

Over the past decade, CRISPR/Cas-based gene editing has become a powerful tool for generating mutations in a variety of model organisms, from Escherichia coli to zebrafish, rodents and large mammals. CRISPR/Cas-based gene editing effectively generates insertions or deletions (indels), which allow for rapid gene disruption. However, a large proportion of human genetic diseases are caused by single-base-pair substitutions, which result in more subtle alterations to protein function, and which require more complex and precise editing to recreate in model systems. Precise genome editing (PGE) methods, however, typically have efficiencies of less than a tenth of those that generate less-specific indels, and so there has been a great deal of effort to improve PGE efficiency. Such optimisations include optimal guide RNA and mutation-bearing donor DNA template design, modulation of DNA repair pathways that underpin how edits result from Cas-induced cuts, and the development of Cas9 fusion proteins that introduce edits via alternative mechanisms. In this Review, we provide an overview of the recent progress in optimising PGE methods and their potential for generating models of human genetic disease.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Humanos , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Pez Cebra/genética , Proteína 9 Asociada a CRISPR/genética , Mutación/genética , Mamíferos
20.
Nat Commun ; 14(1): 1258, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36878908

RESUMEN

Neural crest cells are highly multipotent stem cells, but it remains unclear how their fate restriction to specific fates occurs. The direct fate restriction model hypothesises that migrating cells maintain full multipotency, whilst progressive fate restriction envisages fully multipotent cells transitioning to partially-restricted intermediates before committing to individual fates. Using zebrafish pigment cell development as a model, we show applying NanoString hybridization single cell transcriptional profiling and RNAscope in situ hybridization that neural crest cells retain broad multipotency throughout migration and even in post-migratory cells in vivo, with no evidence for partially-restricted intermediates. We find that leukocyte tyrosine kinase early expression marks a multipotent stage, with signalling driving iridophore differentiation through repression of fate-specific transcription factors for other fates. We reconcile the direct and progressive fate restriction models by proposing that pigment cell development occurs directly, but dynamically, from a highly multipotent state, consistent with our recently-proposed Cyclical Fate Restriction model.


Asunto(s)
Conducción de Automóvil , Pez Cebra , Animales , Pez Cebra/genética , Células Madre Hematopoyéticas , Células Madre Multipotentes , Diferenciación Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA