Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Dev Dyn ; 251(4): 729-742, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34647658

RESUMEN

BACKGROUND: Cell fate decisions are governed by interactions between sequence-specific transcription factors and a dynamic chromatin landscape. Zebrafish offer a powerful system for probing the mechanisms that drive these cell fate choices, especially in the context of early embryogenesis. However, technical challenges associated with conventional methods for chromatin profiling have slowed progress toward understanding the exact relationships between chromatin changes, transcription factor binding, and cellular differentiation during zebrafish embryogenesis. RESULTS: To overcome these challenges, we adapted the chromatin profiling methods Cleavage Under Targets and Release Using Nuclease (CUT&RUN) and CUT&Tag for use in zebrafish and applied these methods to generate high-resolution enrichment maps for H3K4me3, H3K27me3, H3K9me3, RNA polymerase II, and the histone variant H2A.Z using tissue isolated from whole, mid-gastrula stage embryos. Using this data, we identify a subset of genes that may be bivalently regulated during both zebrafish and mouse gastrulation, provide evidence for an evolving H2A.Z landscape during embryo development, and demonstrate the effectiveness of CUT&RUN for detecting H3K9me3 enrichment at repetitive sequences. CONCLUSIONS: Our results demonstrate the power of combining CUT&RUN and CUT&Tag methods with the strengths of the zebrafish system to define emerging chromatin landscapes in the context of vertebrate embryogenesis.


Asunto(s)
Cromatina , Pez Cebra , Animales , Cromatina/genética , Inmunoprecipitación de Cromatina , Desarrollo Embrionario/genética , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Pez Cebra/genética
2.
Trends Genet ; 35(11): 818-827, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31399242

RESUMEN

Chemical modification of nucleotide bases in DNA provides one mechanism for conveying information in addition to the genetic code. 5-methylcytosine (5mC) represents the most common chemically modified base in eukaryotic genomes. Sometimes referred to simply as DNA methylation, in eukaryotes 5mC is most prevalent at CpG dinucleotides and is frequently associated with transcriptional repression of transposable elements. However, 5mC levels and distributions are variable across phylogenies, and emerging evidence suggests that the functions of DNA methylation may be more diverse and complex than was previously appreciated. We summarize the current understanding of DNA methylation profiles and functions in different eukaryotic lineages.


Asunto(s)
Metilación de ADN , Eucariontes/genética , Regulación de la Expresión Génica , Epigénesis Genética , Genoma , Genómica/métodos
3.
Proc Natl Acad Sci U S A ; 112(9): 2770-5, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25730872

RESUMEN

The intestinal epithelium forms a barrier protecting the organism from microbes and other proinflammatory stimuli. The integrity of this barrier and the proper response to infection requires precise regulation of powerful immune homing signals such as tumor necrosis factor (TNF). Dysregulation of TNF leads to inflammatory bowel diseases (IBD), but the mechanism controlling the expression of this potent cytokine and the events that trigger the onset of chronic inflammation are unknown. Here, we show that loss of function of the epigenetic regulator ubiquitin-like protein containing PHD and RING finger domains 1 (uhrf1) in zebrafish leads to a reduction in tnfa promoter methylation and the induction of tnfa expression in intestinal epithelial cells (IECs). The increase in IEC tnfa levels is microbe-dependent and results in IEC shedding and apoptosis, immune cell recruitment, and barrier dysfunction, consistent with chronic inflammation. Importantly, tnfa knockdown in uhrf1 mutants restores IEC morphology, reduces cell shedding, and improves barrier function. We propose that loss of epigenetic repression and TNF induction in the intestinal epithelium can lead to IBD onset.


Asunto(s)
Metilación de ADN , Epigénesis Genética/fisiología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/embriología , Pez Cebra/embriología , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Inflamación/genética , Inflamación/mortalidad , Inflamación/patología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Transactivadores/genética , Transactivadores/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Methods ; 66(3): 433-40, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23792917

RESUMEN

The Gal4-UAS regulatory system of yeast is widely used to modulate gene expression in Drosophila; however, there are limitations to its usefulness in transgenic zebrafish, owing to progressive methylation and silencing of the CpG-rich multicopy upstream activation sequence. Although a modified, less repetitive UAS construct may overcome this problem, it is highly desirable to have additional transcriptional regulatory systems that can be applied independently or in combination with the Gal4/UAS system for intersectional gene expression. The Q transcriptional regulatory system of Neurospora crassa functions similarly to Gal4/UAS. QF is a transcriptional activator that binds to the QUAS upstream regulatory sequence to drive reporter gene expression. Unlike Gal4, the QF binding site does not contain essential CpG dinucleotide sequences that are subject to DNA methylation. The QS protein is a repressor of QF mediated transcriptional activation akin to Gal80. The functionality of the Q system has been demonstrated in Drosophila and Caenorhabditis elegans and we now report its successful application to a vertebrate model, the zebrafish, Danio rerio. Several tissue-specific promoters were used to drive QF expression in stable transgenic lines, as assessed by activation of a QUAS:GFP transgene. The QS repressor was found to dramatically reduce QF activity in injected zebrafish embryos; however, a similar repression has not yet been achieved in transgenic animals expressing QS under the control of ubiquitous promoters. A dual reporter construct containing both QUAS and UAS, each upstream of different fluorescent proteins was also generated and tested in transient assays, demonstrating that the two systems can work in parallel within the same cell. The adoption of the Q system should greatly increase the versatility and power of transgenic approaches for regulating gene expression in zebrafish.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Ingeniería Genética/métodos , Pez Cebra/genética , Animales , Animales Modificados Genéticamente/metabolismo , Regulación de la Expresión Génica/genética , Genes Fúngicos , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Neurospora crassa/genética , Factores de Transcripción/genética , Activación Transcripcional
5.
bioRxiv ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38496550

RESUMEN

The structural organization of eukaryotic genomes is contingent upon the fractionation of DNA into transcriptionally permissive euchromatin and repressive heterochromatin. However, we have a limited understanding of how these distinct states are first established during animal embryogenesis. Histone 3 lysine 9 trimethylation (H3K9me3) is critical to heterochromatin formation and bulk establishment of this mark is thought to help drive large-scale remodeling of an initially naive chromatin state during animal embryogenesis. However, a detailed understanding of this process is lacking. Here, we leverage CUT&RUN to define the emerging H3K9me3 landscape of the zebrafish embryo with high sensitivity and temporal resolution. Despite the prevalence of DNA transposons in the zebrafish genome, we found that LTR transposons are preferentially targeted for embryonic H3K9me3 deposition, with different families exhibiting distinct establishment timelines. High signal-to-noise ratios afforded by CUT&RUN revealed new, emerging sites of low-amplitude H3K9me3 that initiated before the major wave of zygotic genome activation (ZGA). Early sites of establishment predominated at specific subsets of transposons and were particularly enriched for transposon sequences with maternal piRNAs and pericentromeric localization. Notably, the number of H3K9me3 enriched sites increased linearly across blastula development, while quantitative comparison revealed a >10-fold genome-wide increase in H3K9me3 signal at established sites over just 30 minutes at the onset of ZGA. Continued maturation of the H3K9me3 landscape was observed beyond the initial wave of bulk establishment.

6.
Proc Natl Acad Sci U S A ; 107(19): 8689-94, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20395551

RESUMEN

Cytosine DNA methylation is a heritable epigenetic mark present in many eukaryotic organisms. Although DNA methylation likely has a conserved role in gene silencing, the levels and patterns of DNA methylation appear to vary drastically among different organisms. Here we used shotgun genomic bisulfite sequencing (BS-Seq) to compare DNA methylation in eight diverse plant and animal genomes. We found that patterns of methylation are very similar in flowering plants with methylated cytosines detected in all sequence contexts, whereas CG methylation predominates in animals. Vertebrates have methylation throughout the genome except for CpG islands. Gene body methylation is conserved with clear preference for exons in most organisms. Furthermore, genes appear to be the major target of methylation in Ciona and honey bee. Among the eight organisms, the green alga Chlamydomonas has the most unusual pattern of methylation, having non-CG methylation enriched in exons of genes rather than in repeats and transposons. In addition, the Dnmt1 cofactor Uhrf1 has a conserved function in maintaining CG methylation in both transposons and gene bodies in the mouse, Arabidopsis, and zebrafish genomes.


Asunto(s)
Metilación de ADN/genética , Evolución Molecular , Plantas/genética , Animales , Arabidopsis/genética , Exones/genética , Intrones/genética , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Sistemas de Lectura Abierta/genética , Filogenia , Secuencias Repetitivas de Ácidos Nucleicos/genética , Transactivadores/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
7.
Dev Biol ; 352(2): 191-201, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21223961

RESUMEN

The yeast Gal4/UAS transcriptional activation system is a powerful tool for regulating gene expression in Drosophila and has been increasing in popularity for developmental studies in zebrafish. It is also useful for studying the basis of de novo transcriptional silencing. Fluorescent reporter genes under the control of multiple tandem copies of the upstream activator sequence (UAS) often show evidence of variegated expression and DNA methylation in transgenic zebrafish embryos. To characterize this systematically, we monitored the progression of transcriptional silencing of UAS-regulated transgenes that differ in their integration sites and in the repetitive nature of the UAS. Transgenic larvae were examined in three generations for tissue-specific expression of a green fluorescent protein (GFP) reporter and DNA methylation at the UAS. Single insertions containing four distinct upstream activator sequences were far less susceptible to methylation than insertions containing fourteen copies of the same UAS. In addition, transgenes that integrated in or adjacent to transposon sequence exhibited silencing regardless of the number of UAS sites included in the transgene. Placement of promoter-driven Gal4 upstream of UAS-regulated responder genes in a single bicistronic construct also appeared to accelerate silencing and methylation. The results demonstrate the utility of the zebrafish for efficient tracking of gene silencing mechanisms across several generations, as well as provide useful guidelines for optimal Gal4-regulated gene expression in organisms subject to DNA methylation.


Asunto(s)
Pez Cebra/crecimiento & desarrollo , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Metilación de ADN , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Recombinantes/genética , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Activación Transcripcional
8.
Dev Dyn ; 240(9): 2101-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21805532

RESUMEN

Site-specific recombinases (SSRs) are powerful tools for genome manipulation, used in diverse organisms including Drosophila melanogaster, mouse, Arabidopsis, zebrafish, and human cultured cells. The integrase from the bacteriophage ΦC31 belongs to the large serine family of integrases, and in contrast to other widely used SSRs such as Cre and Flp, recombination is directional and therefore irreversible. We have developed a vector system for recombinase-mediated cassette exchange (RMCE) in the zebrafish, allowing swapping of the coding sequence in an integrated transgene. Utilizing codon-optimized ΦC31 integrase RNA bearing the 3'UTR from the nanos1 gene, we replaced the egfp coding sequence of an integrated reporter transgene with mCherry coding sequence. Recombination was achieved at high efficiency in both somatic cells and in the germline. We demonstrate an effective approach to RMCE, increasing the repertoire of tools available to manipulate the zebrafish genome.


Asunto(s)
Bacteriófagos/enzimología , ADN Nucleotidiltransferasas/metabolismo , Integrasas/metabolismo , Animales , Bacteriófagos/genética , ADN Nucleotidiltransferasas/genética , Células Germinativas , Integrasas/genética , Modelos Biológicos , Pez Cebra
9.
Front Cell Dev Biol ; 10: 832461, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35356281

RESUMEN

Heterochromatin formation and maintenance is critical for the repression of transcription from repetitive sequences. However, in vivo tools for monitoring heterochromatin mediated repression of repeats in the context of vertebrate development have been lacking. Here we demonstrate that a large concatemeric transgene integration containing the dsRed fluorescent reporter under the control of a ubiquitous promoter recapitulates molecular hallmarks of heterochromatic silencing, and that expression from the transgene array can be reactivated by depletion of known regulators of heterochromatin. We then use this reporter to identify a previously unappreciated role for the zebrafish NSD1 orthologs, Nsd1a and Nsd1b, in promoting heterochromatin mediated repression. Our results provide proof-principle that this transgenic reporter line can be used to rapidly identify genes with potential roles in heterochromatic silencing in the context of a live, vertebrate organism.

10.
Dev Biol ; 334(1): 213-23, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19631206

RESUMEN

Developmental mechanisms regulating gene expression and the stable acquisition of cell fate direct cytodifferentiation during organogenesis. Moreover, it is likely that such mechanisms could be exploited to repair or regenerate damaged organs. DNA methyltransferases (Dnmts) are enzymes critical for epigenetic regulation, and are used in concert with histone methylation and acetylation to regulate gene expression and maintain genomic integrity and chromosome structure. We carried out two forward genetic screens for regulators of endodermal organ development. In the first, we screened for altered morphology of developing digestive organs, while in the second we screed for the lack of terminally differentiated cell types in the pancreas and liver. From these screens, we identified two mutant alleles of zebrafish dnmt1. Both lesions are predicted to eliminate dnmt1 function; one is a missense mutation in the catalytic domain and the other is a nonsense mutation that eliminates the catalytic domain. In zebrafish dnmt1 mutants, the pancreas and liver form normally, but begin to degenerate after 84 h post fertilization (hpf). Acinar cells are nearly abolished through apoptosis by 100 hpf, though neither DNA replication, nor entry into mitosis is halted in the absence of detectable Dnmt1. However, endocrine cells and ducts are largely spared. Surprisingly, dnmt1 mutants and dnmt1 morpholino-injected larvae show increased capacity for pancreatic beta cell regeneration in an inducible model of pancreatic beta cell ablation. Thus, our data suggest that Dnmt1 is dispensable for pancreatic duct or endocrine cell formation, but not for acinar cell survival. In addition, Dnmt1 may influence the differentiation of pancreatic beta cell progenitors or the reprogramming of cells toward the pancreatic beta cell fate.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Páncreas/citología , Regeneración/fisiología , Proteínas de Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Supervivencia Celular , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Células Endocrinas/metabolismo , Técnica del Anticuerpo Fluorescente , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Datos de Secuencia Molecular , Páncreas/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
11.
Genetics ; 182(3): 747-55, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19433629

RESUMEN

Epigenetic regulation of transcriptional silencing is essential for normal development. Despite its importance, in vivo systems for examining gene silencing at cellular resolution have been lacking in developing vertebrates. We describe a transgenic approach that allows monitoring of an epigenetically regulated fluorescent reporter in developing zebrafish and their progeny. Using a self-reporting Gal4-VP16 gene/enhancer trap vector, we isolated tissue-specific drivers that regulate expression of the green fluorescent protein (GFP) gene through a multicopy, upstream activator sequence (UAS). Transgenic larvae initially exhibit robust fluorescence (GFP(high)); however, in subsequent generations, gfp expression is mosaic (GFP(low)) or entirely absent (GFP(off)), despite continued Gal4-VP16 activity. We find that transcriptional repression is heritable and correlated with methylation of the multicopy UAS. Silenced transgenes can be reactivated by increasing Gal4-VP16 levels or in DNA methyltransferase-1 (dnmt1) mutants. Strikingly, in dnmt1 homozygous mutants, reactivation of gfp expression occurs in a reproducible subset of cells, raising the possibility of different sensitivities or alternative silencing mechanisms in discrete cell populations. The results demonstrate the power of the zebrafish system for in vivo monitoring of epigenetic processes using a genetic approach.


Asunto(s)
Silenciador del Gen , Proteínas Fluorescentes Verdes/genética , Activación Transcripcional , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Epigénesis Genética , Femenino , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Larva/genética , Larva/metabolismo , Masculino , Microscopía Fluorescente , Mutación , Transactivadores/genética , Transactivadores/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
12.
F1000Res ; 92020.
Artículo en Inglés | MEDLINE | ID: mdl-32528656

RESUMEN

Early animal development is characterized by intense reorganization of the embryonic genome, including large-scale changes in chromatin structure and in the DNA and histone modifications that help shape this structure. Particularly profound shifts in the chromatin landscape are associated with the maternal-to-zygotic transition, when the zygotic genome is first transcribed and maternally loaded transcripts are degraded. The accessibility of the early zebrafish embryo facilitates the interrogation of chromatin during this critical window of development, making it an important model for early chromatin regulation. Here, we review our current understanding of chromatin dynamics during early zebrafish development, highlighting new advances as well as similarities and differences between early chromatin regulation in zebrafish and other species.


Asunto(s)
Cromatina/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteínas de Pez Cebra , Pez Cebra/embriología , Animales , Cigoto
13.
Nat Commun ; 10(1): 1551, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948728

RESUMEN

The segregation of eukaryotic genomes into euchromatin and heterochromatin represents a fundamental and poorly understood process. Here, we demonstrate that genome-wide establishment of heterochromatin is triggered by the maternal to zygotic transition (MZT) during zebrafish embryogenesis. We find that prior to MZT, zebrafish lack hallmarks of heterochromatin including histone H3 lysine 9 trimethylation (H3K9me3) and condensed chromatin ultrastructure. Global establishment of heterochromatic features occurs following MZT and requires both activation of the zygotic genome and degradation of maternally deposited RNA. Mechanistically, we demonstrate that zygotic transcription of the micro RNA miR-430 promotes degradation of maternal RNA encoding the chromatin remodeling protein Smarca2, and that clearance of Smarca2 is required for global heterochromatin establishment in the early embryo. Our results identify MZT as a key developmental regulator of heterochromatin establishment during vertebrate embryogenesis and uncover functions for Smarca2 in protecting the embryonic genome against heterochromatinization.


Asunto(s)
Desarrollo Embrionario/genética , Heterocromatina/genética , Pez Cebra/embriología , Animales , Cromatina/genética , Cromatina/metabolismo , Cromatina/ultraestructura , Embrión no Mamífero/citología , Regulación del Desarrollo de la Expresión Génica , Heterocromatina/metabolismo , Heterocromatina/ultraestructura , MicroARNs/metabolismo , MicroARNs/fisiología , Transcripción Genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología
14.
Elife ; 82019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31513014

RESUMEN

In many vertebrates, establishment of Left-Right (LR) asymmetry results from the activity of a ciliated organ functioning as the LR Organizer (LRO). While regulation of the formation of this structure by major signaling pathways has been described, the transcriptional control of LRO formation is poorly understood. Using the zebrafish model, we show that the transcription factors and cofactors mediating or regulating the transcriptional outcome of the Hippo signaling pathway play a pivotal role in controlling the expression of genes essential to the formation of the LRO including ligands and receptors of signaling pathways involved in this process and most genes required for motile ciliogenesis. Moreover, the transcription cofactor, Vgll4l regulates epigenetic programming in LRO progenitors by controlling the expression of writers and readers of DNA methylation marks. Altogether, our study uncovers a novel and essential role for the transcriptional effectors and regulators of the Hippo pathway in establishing LR asymmetry.


Asunto(s)
Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Factores de Transcripción/metabolismo , Pez Cebra/anatomía & histología , Pez Cebra/embriología , Animales
15.
Cell Rep ; 26(3): 720-732.e4, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650362

RESUMEN

Ten-eleven translocation (Tet) enzymes (Tet1/2/3) mediate 5-methylcytosine (5mC) hydroxylation, which can facilitate DNA demethylation and thereby impact gene expression. Studied mostly for how mutant isoforms impact cancer, the normal roles for Tet enzymes during organogenesis are largely unknown. By analyzing compound mutant zebrafish, we discovered a requirement for Tet2/3 activity in the embryonic heart for recruitment of epicardial progenitors, associated with development of the atrial-ventricular canal (AVC). Through a combination of methylation, hydroxymethylation, and transcript profiling, the genes encoding the activin A subunit Inhbaa (in endocardium) and Sox9b (in myocardium) were implicated as demethylation targets of Tet2/3 and critical for organization of AVC-localized extracellular matrix (ECM), facilitating migration of epicardial progenitors onto the developing heart tube. This study elucidates essential DNA demethylation modifications that govern gene expression changes during cardiac development with striking temporal and lineage specificities, highlighting complex interactions in multiple cell populations during development of the vertebrate heart.


Asunto(s)
Dioxigenasas/genética , Matriz Extracelular/metabolismo , Corazón/fisiopatología , Organogénesis/genética , Proteínas de Pez Cebra/genética , Animales , Movimiento Celular , Pez Cebra
16.
Elife ; 72018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30484769

RESUMEN

Pericentromeric satellite repeats are enriched in 5-methylcytosine (5mC). Loss of 5mC at these sequences is common in cancer and is a hallmark of Immunodeficiency, Centromere and Facial abnormalities (ICF) syndrome. While the general importance of 5mC is well-established, the specific functions of 5mC at pericentromeres are less clear. To address this deficiency, we generated a viable animal model of pericentromeric hypomethylation through mutation of the ICF-gene ZBTB24. Deletion of zebrafish zbtb24 caused a progressive loss of 5mC at pericentromeres and ICF-like phenotypes. Hypomethylation of these repeats triggered derepression of pericentromeric transcripts and activation of an interferon-based innate immune response. Injection of pericentromeric RNA is sufficient to elicit this response in wild-type embryos, and mutation of the MDA5-MAVS dsRNA-sensing machinery blocks the response in mutants. These findings identify activation of the innate immune system as an early consequence of pericentromeric hypomethylation, implicating derepression of pericentromeric transcripts as a trigger of autoimmunity. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Asunto(s)
Centrómero , Metilación de ADN , Cara/anomalías , Síndromes de Inmunodeficiencia/patología , Interferones/metabolismo , Animales , Modelos Animales de Enfermedad , Cara/patología , Técnicas de Inactivación de Genes , Inmunidad Innata , Enfermedades de Inmunodeficiencia Primaria , Pez Cebra
17.
Elife ; 72018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30325306

RESUMEN

TET enzymes convert 5-methylcytosine to 5-hydroxymethylcytosine and higher oxidized derivatives. TETs stably associate with and are post-translationally modified by the nutrient-sensing enzyme OGT, suggesting a connection between metabolism and the epigenome. Here, we show for the first time that modification by OGT enhances TET1 activity in vitro. We identify a TET1 domain that is necessary and sufficient for binding to OGT and report a point mutation that disrupts the TET1-OGT interaction. We show that this interaction is necessary for TET1 to rescue hematopoetic stem cell production in tet mutant zebrafish embryos, suggesting that OGT promotes TET1's function during development. Finally, we show that disrupting the TET1-OGT interaction in mouse embryonic stem cells changes the abundance of TET2 and 5-methylcytosine, which is accompanied by alterations in gene expression. These results link metabolism and epigenetic control, which may be relevant to the developmental and disease processes regulated by these two enzymes.


Asunto(s)
Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Células Madre Embrionarias de Ratones/metabolismo , N-Acetilglucosaminiltransferasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Pez Cebra/genética , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Metilación de ADN/genética , Epigenómica , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/genética , Humanos , Ratones , Mutación , Unión Proteica/genética , Dominios Proteicos/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
18.
Cell Rep ; 12(7): 1133-43, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26257178

RESUMEN

The Tet family of methylcytosine dioxygenases (Tet1, Tet2, and Tet3) convert 5-methylcytosine to 5-hydroxymethylcytosine. To date, functional overlap among Tet family members has not been examined systematically in the context of embryonic development. To clarify the potential for overlap among Tet enzymes during development, we mutated the zebrafish orthologs of Tet1, Tet2, and Tet3 and examined single-, double-, and triple-mutant genotypes. Here, we identify Tet2 and Tet3 as the major 5-methylcytosine dioxygenases in the zebrafish embryo and uncover a combined requirement for Tet2 and Tet3 in hematopoietic stem cell (HSC) emergence. We demonstrate that Notch signaling in the hemogenic endothelium is regulated by Tet2/3 prior to HSC emergence and show that restoring expression of the downstream gata2b/scl/runx1 transcriptional network can rescue HSCs in tet2/3 double mutant larvae. Our results reveal essential, overlapping functions for tet genes during embryonic development and uncover a requirement for 5hmC in regulating HSC production.


Asunto(s)
Dioxigenasas/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Dioxigenasas/genética , Desarrollo Embrionario , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética
19.
Nat Cell Biol ; 15(6): 591-601, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23644468

RESUMEN

The transition zone is a specialized compartment found at the base of cilia, adjacent to the centriole distal end, where axonemal microtubules are heavily crosslinked to the surrounding membrane to form a barrier that gates the ciliary compartment. A number of ciliopathy molecules have been found to associate with the transition zone, but factors that directly recognize axonemal microtubules to specify transition zone assembly at the cilia base remain unclear. Here, through quantitative centrosome proteomics, we identify an axoneme-associated protein, CEP162 (KIAA1009), tethered specifically at centriole distal ends to promote transition zone assembly. CEP162 interacts with core transition zone components, and mediates their association with microtubules. Loss of CEP162 arrests ciliogenesis at the stage of transition zone assembly. Abolishing its centriolar tethering, however, allows CEP162 to stay on the growing end of the axoneme and ectopically assemble transition zone components at cilia tips. This generates extra-long cilia with strikingly swollen tips that actively release ciliary contents into the extracellular environment. CEP162 is thus an axoneme-recognition protein pre-tethered at centriole distal ends before ciliogenesis to promote and restrict transition zone formation specifically at the cilia base.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Antígenos de Neoplasias/metabolismo , Axonema/metabolismo , Centriolos/metabolismo , Cilios/metabolismo , Proteínas de Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Células 3T3 , Adenosina Trifosfatasas/genética , Animales , Antígenos de Neoplasias/genética , Proteínas de Ciclo Celular , Línea Celular , Centrosoma/metabolismo , Proteínas del Citoesqueleto , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/metabolismo , Proteínas de Neoplasias/genética , Proteómica , Interferencia de ARN , ARN Interferente Pequeño
20.
Prog Mol Biol Transl Sci ; 101: 193-218, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21507352

RESUMEN

DNA methylation is crucial for normal development and cellular differentiation in many large-genome eukaryotes. The small tropical freshwater fish Danio rerio (zebrafish) has recently emerged as a powerful system for the study of DNA methylation, especially in the context of development. This review summarizes our current knowledge of DNA methylation in zebrafish and provides evidence for the general conservation of this system with mammals. In addition, emerging strategies are highlighted that use the fish model to address some of the key unanswered questions in DNA methylation research.


Asunto(s)
Metilación de ADN , Pez Cebra/genética , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA