Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Vis Exp ; (207)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38856229

RESUMEN

The mammalian heart is a complex organ formed during development via highly diverse populations of progenitor cells. The origin, timing of recruitment, and fate of these progenitors are vital for the proper development of this organ. The molecular mechanisms that govern the morphogenesis of the heart are essential for understanding the pathogenesis of congenital heart diseases and embryonic cardiac regeneration. Classical approaches to investigate these mechanisms employed the generation of transgenic mice to assess the function of specific genes during cardiac development. However, mouse transgenesis is a complex, time-consuming process that often cannot be performed to assess the role of specific genes during heart development. To address this, we have developed a protocol for efficient electroporation and culture of mouse embryonic hearts, enabling transient transgenesis to rapidly assess the effect of gain- or loss-of-function of genes involved in cardiac development. Using this methodology, we successfully overexpressed Meis1 in the embryonic heart, with a preference for epicardial cell transfection, demonstrating the capabilities of the technique.


Asunto(s)
Electroporación , Técnicas de Transferencia de Gen , Corazón , Animales , Ratones , Corazón/embriología , Electroporación/métodos , Ratones Transgénicos , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Embarazo
2.
Curr Top Dev Biol ; 159: 406-437, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38729683

RESUMEN

Transcriptional regulation plays a pivotal role in orchestrating the intricate genetic programs governing embryonic development. The expression of developmental genes relies on the combined activity of several cis-regulatory elements (CREs), such as enhancers and silencers, which can be located at long linear distances from the genes that they regulate and that interact with them through establishment of chromatin loops. Mutations affecting their activity or interaction with their target genes can lead to developmental disorders and are thought to have importantly contributed to the evolution of the animal body plan. The income of next-generation-sequencing approaches has allowed identifying over a million of sequences with putative regulatory potential in the human genome. Characterizing their function and establishing gene-CREs maps is essential to decode the logic governing developmental gene expression and is one of the major challenges of the post-genomic era. Chromatin 3D organization plays an essential role in determining how CREs specifically contact their target genes while avoiding deleterious off-target interactions. Our understanding of these aspects has greatly advanced with the income of chromatin conformation capture techniques and fluorescence microscopy approaches to visualize the organization of DNA elements in the nucleus. Here we will summarize relevant aspects of how the interplay between CRE activity and chromatin 3D organization regulates developmental gene expression and how it relates to pathological conditions and the evolution of animal body plan.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Humanos , Cromatina/metabolismo , Cromatina/genética , Animales , Evolución Molecular
3.
Nat Commun ; 14(1): 8481, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38123574

RESUMEN

The risk of developing drug addiction is strongly influenced by the epigenetic landscape and chromatin remodeling. While histone modifications such as methylation and acetylation have been studied in the ventral tegmental area and nucleus accumbens (NAc), the role of H2A monoubiquitination remains unknown. Our investigations, initially focused on the scaffold protein melanoma-associated antigen D1 (Maged1), reveal that H2A monoubiquitination in the paraventricular thalamus (PVT) significantly contributes to cocaine-adaptive behaviors and transcriptional repression induced by cocaine. Chronic cocaine use increases H2A monoubiquitination, regulated by Maged1 and its partner USP7. Accordingly, Maged1 specific inactivation in thalamic Vglut2 neurons, or USP7 inhibition, blocks cocaine-evoked H2A monoubiquitination and cocaine locomotor sensitization. Additionally, genetic variations in MAGED1 and USP7 are linked to altered susceptibility to cocaine addiction and cocaine-associated symptoms in humans. These findings unveil an epigenetic modification in a non-canonical reward pathway of the brain and a potent marker of epigenetic risk factors for drug addiction in humans.


Asunto(s)
Trastornos Relacionados con Cocaína , Cocaína , Trastornos Relacionados con Sustancias , Humanos , Peptidasa Específica de Ubiquitina 7/metabolismo , Cocaína/farmacología , Cocaína/metabolismo , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/metabolismo , Trastornos Relacionados con Sustancias/genética , Epigénesis Genética , Núcleo Accumbens/metabolismo , Tálamo/metabolismo
4.
Sci Adv ; 8(51): eabo0694, 2022 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-36563144

RESUMEN

The molecular mechanisms that maintain cellular identities and prevent dedifferentiation or transdifferentiation remain mysterious. However, both processes are transiently used during animal regeneration. Therefore, organisms that regenerate their organs, appendages, or even their whole body offer a fruitful paradigm to investigate the regulation of cell fate stability. Here, we used Hydra as a model system and show that Zic4, whose expression is controlled by Wnt3/ß-catenin signaling and the Sp5 transcription factor, plays a key role in tentacle formation and tentacle maintenance. Reducing Zic4 expression suffices to induce transdifferentiation of tentacle epithelial cells into foot epithelial cells. This switch requires the reentry of tentacle battery cells into the cell cycle without cell division and is accompanied by degeneration of nematocytes embedded in these cells. These results indicate that maintenance of cell fate by a Wnt-controlled mechanism is a key process both during homeostasis and during regeneration.

5.
Development ; 149(12)2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35770682

RESUMEN

Modifications in gene regulation are driving forces in the evolution of organisms. Part of these changes involve cis-regulatory elements (CREs), which contact their target genes through higher-order chromatin structures. However, how such architectures and variations in CREs contribute to transcriptional evolvability remains elusive. We use Hoxd genes as a paradigm for the emergence of regulatory innovations, as many relevant enhancers are located in a regulatory landscape highly conserved in amniotes. Here, we analysed their regulation in murine vibrissae and chicken feather primordia, two skin appendages expressing different Hoxd gene subsets, and compared the regulation of these genes in these appendages with that in the elongation of the posterior trunk. In the two former structures, distinct subsets of Hoxd genes are contacted by different lineage-specific enhancers, probably as a result of using an ancestral chromatin topology as an evolutionary playground, whereas the gene regulation that occurs in the mouse and chicken embryonic trunk partially relies on conserved CREs. A high proportion of these non-coding sequences active in the trunk have functionally diverged between species, suggesting that transcriptional robustness is maintained, despite considerable divergence in enhancer sequences.


Asunto(s)
Pollos , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Pollos/genética , Cromatina/genética , Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Secuencias Reguladoras de Ácidos Nucleicos/genética
6.
Genes Dev ; 35(21-22): 1490-1509, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34711654

RESUMEN

Mammalian Hox gene clusters contain a range of CTCF binding sites. In addition to their importance in organizing a TAD border, which isolates the most posterior genes from the rest of the cluster, the positions and orientations of these sites suggest that CTCF may be instrumental in the selection of various subsets of contiguous genes, which are targets of distinct remote enhancers located in the flanking regulatory landscapes. We examined this possibility by producing an allelic series of cumulative in cis mutations in these sites, up to the abrogation of CTCF binding in the five sites located on one side of the TAD border. In the most impactful alleles, the global chromatin architecture of the locus was modified, yet not drastically, illustrating that CTCF sites located on one side of a strong TAD border are sufficient to organize at least part of this insulation. Spatial colinearity in the expression of these genes along the major body axis was nevertheless maintained, despite abnormal expression boundaries. In contrast, strong effects were scored in the selection of target genes responding to particular enhancers, leading to the misregulation of Hoxd genes in specific structures. Altogether, while most enhancer-promoter interactions can occur in the absence of this series of CTCF sites, the binding of CTCF in the Hox cluster is required to properly transform a rather unprecise process into a highly discriminative mechanism of interactions, which is translated into various patterns of transcription accompanied by the distinctive chromatin topology found at this locus. Our allelic series also allowed us to reveal the distinct functional contributions for CTCF sites within this Hox cluster, some acting as insulator elements, others being necessary to anchor or stabilize enhancer-promoter interactions, and some doing both, whereas they all together contribute to the formation of a TAD border. This variety of tasks may explain the amazing evolutionary conservation in the distribution of these sites among paralogous Hox clusters or between various vertebrates.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Animales , Sitios de Unión , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Elementos de Facilitación Genéticos/genética , Genes Homeobox/genética , Mamíferos/genética , Ratones , Mutagénesis
7.
Dev Dyn ; 250(9): 1280-1299, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33497014

RESUMEN

BACKGROUND: During tetrapod limb development, the HOXA13 and HOXD13 transcription factors are critical for the emergence and organization of the autopod, the most distal aspect where digits will develop. Since previous work had suggested that the Dbx2 gene is a target of these factors, we set up to analyze in detail this potential regulatory interaction. RESULTS: We show that HOX13 proteins bind to mammalian-specific sequences at the vicinity of the Dbx2 locus that have enhancer activity in developing digits. However, the functional inactivation of the DBX2 protein did not elicit any particular phenotype related to Hox genes inactivation in digits, suggesting either redundant or compensatory mechanisms. We report that the neighboring Nell2 and Ano6 genes are also expressed in distal limb buds and are in part controlled by the same Dbx2 enhancers despite being localized into two different topologically associating domains (TADs) flanking the Dbx2 locus. CONCLUSIONS: We conclude that Hoxa13 and Hoxd genes cooperatively activate Dbx2 expression in developing digits through binding to mammalian specific regulatory sequences in the Dbx2 neighborhood. Furthermore, these enhancers can overcome TAD boundaries in either direction to co-regulate a set of genes located in distinct chromatin domains.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Animales , Extremidades , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Esbozos de los Miembros/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(49): 31231-31241, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229569

RESUMEN

The HoxD gene cluster is critical for proper limb formation in tetrapods. In the emerging limb buds, different subgroups of Hoxd genes respond first to a proximal regulatory signal, then to a distal signal that organizes digits. These two regulations are exclusive from one another and emanate from two distinct topologically associating domains (TADs) flanking HoxD, both containing a range of appropriate enhancer sequences. The telomeric TAD (T-DOM) contains several enhancers active in presumptive forearm cells and is divided into two sub-TADs separated by a CTCF-rich boundary, which defines two regulatory submodules. To understand the importance of this particular regulatory topology to control Hoxd gene transcription in time and space, we either deleted or inverted this sub-TAD boundary, eliminated the CTCF binding sites, or inverted the entire T-DOM to exchange the respective positions of the two sub-TADs. The effects of such perturbations on the transcriptional regulation of Hoxd genes illustrate the requirement of this regulatory topology for the precise timing of gene activation. However, the spatial distribution of transcripts was eventually resumed, showing that the presence of enhancer sequences, rather than either their exact topology or a particular chromatin architecture, is the key factor. We also show that the affinity of enhancers to find their natural target genes can overcome the presence of both a strong TAD border and an unfavorable orientation of CTCF sites.


Asunto(s)
Factor de Unión a CCCTC/genética , Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos/genética , Genes Homeobox/genética , Animales , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Extremidades/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/genética , Esbozos de los Miembros/crecimiento & desarrollo , Ratones
9.
Nat Commun ; 10(1): 312, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30659200

RESUMEN

Polyps of the cnidarian Hydra maintain their adult anatomy through two developmental organizers, the head organizer located apically and the foot organizer basally. The head organizer is made of two antagonistic cross-reacting components, an activator, driving apical differentiation and an inhibitor, preventing ectopic head formation. Here we characterize the head inhibitor by comparing planarian genes down-regulated when ß-catenin is silenced to Hydra genes displaying a graded apical-to-basal expression and an up-regulation during head regeneration. We identify Sp5 as a transcription factor that fulfills the head inhibitor properties: leading to a robust multiheaded phenotype when knocked-down in Hydra, acting as a transcriptional repressor of Wnt3 and positively regulated by Wnt/ß-catenin signaling. Hydra and zebrafish Sp5 repress Wnt3 promoter activity while Hydra Sp5 also activates its own expression, likely via ß-catenin/TCF interaction. This work identifies Sp5 as a potent feedback loop inhibitor of Wnt/ß-catenin signaling, a function conserved across eumetazoan evolution.


Asunto(s)
Hydra/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Wnt3/genética , beta Catenina/genética , Animales , Evolución Biológica , Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Cabeza/crecimiento & desarrollo , Cabeza/fisiología , Hydra/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Planarias/genética , Interferencia de ARN , Regeneración/fisiología , Transducción de Señal , Proteína Wnt3/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , beta Catenina/metabolismo
10.
PLoS Biol ; 16(11): e3000004, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30475793

RESUMEN

In all tetrapods examined thus far, the development and patterning of limbs require the activation of gene members of the HoxD cluster. In mammals, they are regulated by a complex bimodal process that controls first the proximal patterning and then the distal structure. During the shift from the former to the latter regulation, this bimodal regulatory mechanism allows the production of a domain with low Hoxd gene expression, at which both telomeric (T-DOM) and centromeric regulatory domains (C-DOM) are silent. These cells generate the future wrist and ankle articulations. We analyzed the implementation of this regulatory mechanism in chicken, i.e., in an animal for which large morphological differences exist between fore- and hindlimbs. We report that although this bimodal regulation is globally conserved between the mouse and the chick, some important modifications evolved at least between these two model systems, in particular regarding the activity of specific enhancers, the width of the TAD boundary separating the two regulations, and the comparison between the forelimb versus hindlimb regulatory controls. At least one aspect of these regulations seems to be more conserved between chick and bats than with mouse, which may relate to the extent to which forelimbs and hindlimbs of these various animals differ in their morphologies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox/genética , Genes Homeobox/fisiología , Animales , Embrión de Pollo , Pollos/genética , Elementos de Facilitación Genéticos/genética , Extremidades/embriología , Extremidades/fisiología , Miembro Anterior/embriología , Miembro Posterior/embriología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones/embriología , Ratones/genética , Ratones Endogámicos C57BL , Organogénesis , Transcripción Genética/genética
11.
Nature ; 562(7726): 272-276, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30283134

RESUMEN

The emergence of multiple axes is an essential element in the establishment of the mammalian body plan. This process takes place shortly after implantation of the embryo within the uterus and relies on the activity of gene regulatory networks that coordinate transcription in space and time. Whereas genetic approaches have revealed important aspects of these processes1, a mechanistic understanding is hampered by the poor experimental accessibility of early post-implantation stages. Here we show that small aggregates of mouse embryonic stem cells (ESCs), when stimulated to undergo gastrulation-like events and elongation in vitro, can organize a post-occipital pattern of neural, mesodermal and endodermal derivatives that mimic embryonic spatial and temporal gene expression. The establishment of the three major body axes in these 'gastruloids'2,3 suggests that the mechanisms involved are interdependent. Specifically, gastruloids display the hallmarks of axial gene regulatory systems as exemplified by the implementation of collinear Hox transcriptional patterns along an extending antero-posterior axis. These results reveal an unanticipated self-organizing capacity of aggregated ESCs and suggest that gastruloids could be used as a complementary system to study early developmental events in the mammalian embryo.


Asunto(s)
Tipificación del Cuerpo , Gástrula/citología , Gástrula/embriología , Células Madre Embrionarias de Ratones/citología , Organoides/citología , Organoides/embriología , Animales , Tipificación del Cuerpo/genética , Gástrula/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/genética , Técnicas In Vitro , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Organoides/metabolismo , Factores de Tiempo
12.
Genes Dev ; 31(22): 2264-2281, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29273679

RESUMEN

The mammalian HoxD cluster lies between two topologically associating domains (TADs) matching distinct enhancer-rich regulatory landscapes. During limb development, the telomeric TAD controls the early transcription of Hoxd genes in forearm cells, whereas the centromeric TAD subsequently regulates more posterior Hoxd genes in digit cells. Therefore, the TAD boundary prevents the terminal Hoxd13 gene from responding to forearm enhancers, thereby allowing proper limb patterning. To assess the nature and function of this CTCF-rich DNA region in embryos, we compared chromatin interaction profiles between proximal and distal limb bud cells isolated from mutant stocks where various parts of this boundary region were removed. The resulting progressive release in boundary effect triggered inter-TAD contacts, favored by the activity of the newly accessed enhancers. However, the boundary was highly resilient, and only a 400-kb deletion, including the whole-gene cluster, was eventually able to merge the neighboring TADs into a single structure. In this unified TAD, both proximal and distal limb enhancers nevertheless continued to work independently over a targeted transgenic reporter construct. We propose that the whole HoxD cluster is a dynamic TAD border and that the exact boundary position varies depending on both the transcriptional status and the developmental context.


Asunto(s)
Genes Homeobox , Familia de Multigenes , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Elementos de Facilitación Genéticos , Esbozos de los Miembros/metabolismo , Ratones , Eliminación de Secuencia , Transcripción Genética , Cohesinas
13.
Genome Biol ; 18(1): 149, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28784160

RESUMEN

BACKGROUND: The transcriptional activation of HoxD genes during mammalian limb development involves dynamic interactions with two topologically associating domains (TADs) flanking the HoxD cluster. In particular, the activation of the most posterior HoxD genes in developing digits is controlled by regulatory elements located in the centromeric TAD (C-DOM) through long-range contacts. RESULTS: To assess the structure-function relationships underlying such interactions, we measured compaction levels and TAD discreteness using a combination of chromosome conformation capture (4C-seq) and DNA FISH. We assessed the robustness of the TAD architecture by using a series of genomic deletions and inversions that impact the integrity of this chromatin domain and that remodel long-range contacts. We report multi-partite associations between HoxD genes and up to three enhancers. We find that the loss of native chromatin topology leads to the remodeling of TAD structure following distinct parameters. CONCLUSIONS: Our results reveal that the recomposition of TAD architectures after large genomic re-arrangements is dependent on a boundary-selection mechanism in which CTCF mediates the gating of long-range contacts in combination with genomic distance and sequence specificity. Accordingly, the building of a recomposed TAD at this locus depends on distinct functional and constitutive parameters.


Asunto(s)
Genes Homeobox , Sitios Genéticos , Genómica , Animales , Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Reordenamiento Génico , Islas Genómicas , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Hibridación Fluorescente in Situ , Ratones , Familia de Multigenes , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética
14.
Proc Natl Acad Sci U S A ; 113(48): E7720-E7729, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27856734

RESUMEN

Vertebrate Hox genes encode transcription factors operating during the development of multiple organs and structures. However, the evolutionary mechanism underlying this remarkable pleiotropy remains to be fully understood. Here, we show that Hoxd8 and Hoxd9, two genes of the HoxD complex, are transcribed during mammary bud (MB) development. However, unlike in other developmental contexts, their coexpression does not rely on the same regulatory mechanism. Hoxd8 is regulated by the combined activity of closely located sequences and the most distant telomeric gene desert. On the other hand, Hoxd9 is controlled by an enhancer-rich region that is also located within the telomeric gene desert but has no impact on Hoxd8 transcription, thus constituting an exception to the global regulatory logic systematically observed at this locus. The latter DNA region is also involved in Hoxd gene regulation in other contexts and strongly interacts with Hoxd9 in all tissues analyzed thus far, indicating that its regulatory activity was already operational before the appearance of mammary glands. Within this DNA region and neighboring a strong limb enhancer, we identified a short sequence conserved in therian mammals and capable of enhancer activity in the MBs. We propose that Hoxd gene regulation in embryonic MBs evolved by hijacking a preexisting regulatory landscape that was already at work before the emergence of mammals in structures such as the limbs or the intestinal tract.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Animales , Elementos de Facilitación Genéticos , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Análisis de Secuencia de ARN , Transcripción Genética
15.
Genes Dev ; 30(10): 1172-86, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27198226

RESUMEN

During vertebrate limb development, Hoxd genes are regulated following a bimodal strategy involving two topologically associating domains (TADs) located on either side of the gene cluster. These regulatory landscapes alternatively control different subsets of Hoxd targets, first into the arm and subsequently into the digits. We studied the transition between these two global regulations, a switch that correlates with the positioning of the wrist, which articulates these two main limb segments. We show that the HOX13 proteins themselves help switch off the telomeric TAD, likely through a global repressive mechanism. At the same time, they directly interact with distal enhancers to sustain the activity of the centromeric TAD, thus explaining both the sequential and exclusive operating processes of these two regulatory domains. We propose a model in which the activation of Hox13 gene expression in distal limb cells both interrupts the proximal Hox gene regulation and re-enforces the distal regulation. In the absence of HOX13 proteins, a proximal limb structure grows without any sign of wrist articulation, likely related to an ancestral fish-like condition.


Asunto(s)
Tipificación del Cuerpo/genética , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox/genética , Proteínas de Homeodominio/metabolismo , Dominios Proteicos/genética , Animales , Embrión de Pollo , Elementos de Facilitación Genéticos/genética , Proteínas de Homeodominio/genética , Deformidades Congénitas de las Extremidades/genética , Ratones , Ratones Transgénicos , Mutación , Unión Proteica/genética
16.
J Biol Chem ; 290(45): 26927-26942, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26378230

RESUMEN

A well integrated and hierarchically organized gene regulatory network is responsible for the progressive specification of the forebrain. The transcription factor Six3 is one of the central components of this network. As such, Six3 regulates several components of the network, but its upstream regulators are still poorly characterized. Here we have systematically identified such regulators, taking advantage of the detailed functional characterization of the regulatory region of the medaka fish Six3.2 ortholog and of a time/cost-effective trans-regulatory screening, which complemented and overcame the limitations of in silico prediction approaches. The candidates resulting from this search were validated with dose-response luciferase assays and expression pattern criteria. Reconfirmed candidates with a matching expression pattern were also tested with chromatin immunoprecipitation and functional studies. Our results confirm the previously proposed direct regulation of Pax6 and further demonstrate that Msx2 and Pbx1 are bona fide direct regulators of early Six3.2 distribution in distinct domains of the medaka fish forebrain. They also point to other transcription factors, including Tcf3, as additional regulators of different spatial-temporal domains of Six3.2 expression. The activity of these regulators is discussed in the context of the gene regulatory network proposed for the specification of the forebrain.


Asunto(s)
Proteínas del Ojo/genética , Proteínas de Peces/genética , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/genética , Oryzias/embriología , Oryzias/genética , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Oryzias/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Proteína Homeobox SIX3
17.
Development ; 142(17): 3009-20, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26253404

RESUMEN

Microphthalmos is a rare congenital anomaly characterized by reduced eye size and visual deficits of variable degree. Sporadic and hereditary microphthalmos have been associated with heterozygous mutations in genes fundamental for eye development. Yet, many cases are idiopathic or await the identification of molecular causes. Here we show that haploinsufficiency of Meis1, which encodes a transcription factor with evolutionarily conserved expression in the embryonic trunk, brain and sensory organs, including the eye, causes microphthalmic traits and visual impairment in adult mice. By combining analysis of Meis1 loss-of-function and conditional Meis1 functional rescue with ChIP-seq and RNA-seq approaches we show that, in contrast to its preferential association with Hox-Pbx BSs in the trunk, Meis1 binds to Hox/Pbx-independent sites during optic cup development. In the eye primordium, Meis1 coordinates, in a dose-dependent manner, retinal proliferation and differentiation by regulating genes responsible for human microphthalmia and components of the Notch signaling pathway. In addition, Meis1 is required for eye patterning by controlling a set of eye territory-specific transcription factors, so that in Meis1(-/-) embryos boundaries among the different eye territories are shifted or blurred. We propose that Meis1 is at the core of a genetic network implicated in eye patterning/microphthalmia, and represents an additional candidate for syndromic cases of these ocular malformations.


Asunto(s)
Ojo/embriología , Ojo/metabolismo , Redes Reguladoras de Genes , Proteínas de Homeodominio/metabolismo , Microftalmía/embriología , Microftalmía/genética , Proteínas de Neoplasias/metabolismo , Envejecimiento/patología , Animales , Apoptosis/genética , Secuencia de Bases , Sitios de Unión , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Elementos de Facilitación Genéticos/genética , Haploinsuficiencia/genética , Hematopoyesis/genética , Proteínas de Homeodominio/genética , Humanos , Ratones , Datos de Secuencia Molecular , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Neurogénesis/genética , Unión Proteica , Receptores Notch/metabolismo , Transducción de Señal/genética
18.
Proc Natl Acad Sci U S A ; 112(24): 7542-7, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26034287

RESUMEN

Increasing evidence in the last years indicates that the vast amount of regulatory information contained in mammalian genomes is organized in precise 3D chromatin structures. However, the impact of this spatial chromatin organization on gene expression and its degree of evolutionary conservation is still poorly understood. The Six homeobox genes are essential developmental regulators organized in gene clusters conserved during evolution. Here, we reveal that the Six clusters share a deeply evolutionarily conserved 3D chromatin organization that predates the Cambrian explosion. This chromatin architecture generates two largely independent regulatory landscapes (RLs) contained in two adjacent topological associating domains (TADs). By disrupting the conserved TAD border in one of the zebrafish Six clusters, we demonstrate that this border is critical for preventing competition between promoters and enhancers located in separated RLs, thereby generating different expression patterns in genes located in close genomic proximity. Moreover, evolutionary comparison of Six-associated TAD borders reveals the presence of CCCTC-binding factor (CTCF) sites with diverging orientations in all studied deuterostomes. Genome-wide examination of mammalian HiC data reveals that this conserved CTCF configuration is a general signature of TAD borders, underscoring that common organizational principles underlie TAD compartmentalization in deuterostome evolution.


Asunto(s)
Evolución Molecular , Proteínas Represoras/química , Proteínas Represoras/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Sitios de Unión/genética , Factor de Unión a CCCTC , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Secuencia Conservada , ADN/genética , Elementos de Facilitación Genéticos , Genes Homeobox , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Modelos Genéticos , Familia de Multigenes , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas , Proteínas Represoras/metabolismo , Strongylocentrotus purpuratus/genética , Pez Cebra/genética
19.
Development ; 140(6): 1250-61, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23444355

RESUMEN

The Sox2 transcription factor is active in stem/progenitor cells throughout the developing vertebrate central nervous system. However, its conditional deletion at E12.5 in mouse causes few brain developmental problems, with the exception of the postnatal loss of the hippocampal radial glia stem cells and the dentate gyrus. We deleted Sox2 at E9.5 in the telencephalon, using a Bf1-Cre transgene. We observed embryonic brain defects that were particularly severe in the ventral, as opposed to the dorsal, telencephalon. Important tissue loss, including the medial ganglionic eminence (MGE), was detected at E12.5, causing the subsequent impairment of MGE-derived neurons. The defect was preceded by loss of expression of the essential ventral determinants Nkx2.1 and Shh, and accompanied by ventral spread of dorsal markers. This phenotype is reminiscent of that of mice mutant for the transcription factor Nkx2.1 or for the Shh receptor Smo. Nkx2.1 is known to mediate the initial activation of ventral telencephalic Shh expression. A partial rescue of the normal phenotype at E14.5 was obtained by administration of a Shh agonist. Experiments in Medaka fish indicate that expression of Nkx2.1 is regulated by Sox2 in this species also. We propose that Sox2 contributes to Nkx2.1 expression in early mouse development, thus participating in the region-specific activation of Shh, thereby mediating ventral telencephalic patterning induction.


Asunto(s)
Tipificación del Cuerpo/genética , Desarrollo Embrionario/genética , Proteínas Hedgehog/genética , Proteínas Nucleares/genética , Factores de Transcripción SOXB1/fisiología , Telencéfalo/embriología , Factores de Transcripción/genética , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/metabolismo , Embarazo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Telencéfalo/metabolismo , Factor Nuclear Tiroideo 1 , Factores de Transcripción/metabolismo , Activación Transcripcional/genética
20.
Mech Dev ; 130(2-3): 95-111, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23111324

RESUMEN

The vertebrate forebrain or prosencephalon is patterned at the beginning of neurulation into four major domains: the telencephalic, hypothalamic, retinal and diencephalic anlagen. These domains will then give rise to the majority of the brain structures involved in sensory integration and the control of higher intellectual and homeostatic functions. Understanding how forebrain pattering arises has thus attracted the interest of developmental neurobiologists for decades. As a result, most of its regulators have been identified and their hierarchical relationship is now the object of active investigation. Here, we summarize the main morphogenetic pathways and transcription factors involved in forebrain specification and propose the backbone of a possible gene regulatory network (GRN) governing its specification, taking advantage of the GRN principles elaborated by pioneer studies in simpler organisms. We will also discuss this GRN and its operational logic in the context of the remarkable morphological and functional diversification that the forebrain has undergone during evolution.


Asunto(s)
Redes Reguladoras de Genes , Prosencéfalo/embriología , Animales , Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Humanos , Transducción de Señal , Factores de Transcripción/fisiología , Vertebrados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA