Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Development ; 151(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38828908

RESUMEN

During limb bud formation, axis polarities are established as evidenced by the spatially restricted expression of key regulator genes. In particular, the mutually antagonistic interaction between the GLI3 repressor and HAND2 results in distinct and non-overlapping anterior-distal Gli3 and posterior Hand2 expression domains. This is a hallmark of the establishment of antero-posterior limb axis polarity, together with spatially restricted expression of homeodomain and other transcriptional regulators. Here, we show that TBX3 is required for establishment of the posterior expression boundary of anterior genes in mouse limb buds. ChIP-seq and differential gene expression analysis of wild-type and mutant limb buds identifies TBX3-specific and shared TBX3-HAND2 target genes. High sensitivity fluorescent whole-mount in situ hybridisation shows that the posterior expression boundaries of anterior genes are positioned by TBX3-mediated repression, which excludes anterior genes such as Gli3, Alx4, Hand1 and Irx3/5 from the posterior limb bud mesenchyme. This exclusion delineates the posterior mesenchymal territory competent to establish the Shh-expressing limb bud organiser. In turn, HAND2 is required for Shh activation and cooperates with TBX3 to upregulate shared posterior identity target genes in early limb buds.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Regulación del Desarrollo de la Expresión Génica , Esbozos de los Miembros , Proteínas de Dominio T Box , Animales , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Esbozos de los Miembros/metabolismo , Esbozos de los Miembros/embriología , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteína Gli3 con Dedos de Zinc/metabolismo , Proteína Gli3 con Dedos de Zinc/genética , Regulación hacia Arriba/genética , Tipificación del Cuerpo/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Mesodermo/metabolismo , Mesodermo/embriología
2.
STAR Protoc ; 4(4): 102603, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37742180

RESUMEN

Tissue autofluorescence poses significant challenges for RNA and protein analysis using fluorescence-based techniques. Here, we present a protocol that combines oxidation-mediated autofluorescence reduction with detergent-based tissue permeabilization for whole-mount RNA-fluorescence in situ hybridization (FISH) on mouse embryonic limb buds. We describe the steps for embryo collection, fixation, photochemical bleaching, permeabilization, and RNA-FISH, followed by optical clearing of RNA-FISH and immunofluorescence samples for imaging. The protocol alleviates the need for digital image post-processing to remove autofluorescence and is applicable to other tissues, organs, and vertebrate embryos.


Asunto(s)
Embrión de Mamíferos , ARN , Animales , Ratones , ARN/metabolismo , Hibridación Fluorescente in Situ/métodos , Embrión de Mamíferos/diagnóstico por imagen , Embrión de Mamíferos/metabolismo , Técnica del Anticuerpo Fluorescente
3.
Nat Commun ; 14(1): 3993, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37414772

RESUMEN

A lingering question in developmental biology has centered on how transcription factors with widespread distribution in vertebrate embryos can perform tissue-specific functions. Here, using the murine hindlimb as a model, we investigate the elusive mechanisms whereby PBX TALE homeoproteins, viewed primarily as HOX cofactors, attain context-specific developmental roles despite ubiquitous presence in the embryo. We first demonstrate that mesenchymal-specific loss of PBX1/2 or the transcriptional regulator HAND2 generates similar limb phenotypes. By combining tissue-specific and temporally controlled mutagenesis with multi-omics approaches, we reconstruct a gene regulatory network (GRN) at organismal-level resolution that is collaboratively directed by PBX1/2 and HAND2 interactions in subsets of posterior hindlimb mesenchymal cells. Genome-wide profiling of PBX1 binding across multiple embryonic tissues further reveals that HAND2 interacts with subsets of PBX-bound regions to regulate limb-specific GRNs. Our research elucidates fundamental principles by which promiscuous transcription factors cooperate with cofactors that display domain-restricted localization to instruct tissue-specific developmental programs.


Asunto(s)
Redes Reguladoras de Genes , Factores de Transcripción , Animales , Ratones , Proteínas de Homeodominio/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Development ; 148(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34822715

RESUMEN

SMAD4 regulates gene expression in response to BMP and TGFß signal transduction, and is required for diverse morphogenetic processes, but its target genes have remained largely elusive. Here, we identify the SMAD4 target genes in mouse limb buds using an epitope-tagged Smad4 allele for ChIP-seq analysis in combination with transcription profiling. This analysis shows that SMAD4 predominantly mediates BMP signal transduction during early limb bud development. Unexpectedly, the expression of cholesterol biosynthesis enzymes is precociously downregulated and intracellular cholesterol levels are reduced in Smad4-deficient limb bud mesenchymal progenitors. Most importantly, our analysis reveals a predominant function of SMAD4 in upregulating target genes in the anterior limb bud mesenchyme. Analysis of differentially expressed genes shared between Smad4- and Shh-deficient limb buds corroborates this function of SMAD4 and also reveals the repressive effect of SMAD4 on posterior genes that are upregulated in response to SHH signaling. This analysis uncovers opposing trans-regulatory inputs from SHH- and SMAD4-mediated BMP signal transduction on anterior and posterior gene expression during the digit patterning and outgrowth in early limb buds.


Asunto(s)
Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Hedgehog/metabolismo , Esbozos de los Miembros/embriología , Transducción de Señal , Proteína Smad4/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Miembro Posterior/embriología , Ratones , Ratones Transgénicos , Proteína Smad4/genética
5.
Nat Commun ; 12(1): 5685, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34584102

RESUMEN

Chromatin remodeling and genomic alterations impact spatio-temporal regulation of gene expression, which is central to embryonic development. The analysis of mouse and chicken limb development provides important insights into the morphoregulatory mechanisms, however little is known about the regulatory differences underlying their morphological divergence. Here, we identify the underlying shared and species-specific epigenomic and genomic variations. In mouse forelimb buds, we observe striking synchrony between the temporal dynamics of chromatin accessibility and gene expression, while their divergence in chicken wing buds uncovers species-specific regulatory heterochrony. In silico mapping of transcription factor binding sites and computational footprinting establishes the developmental time-restricted transcription factor-DNA interactions. Finally, the construction of target gene networks for HAND2 and GLI3 transcriptional regulators reveals both conserved and species-specific interactions. Our analysis reveals the impact of genome evolution on the regulatory interactions orchestrating vertebrate limb bud morphogenesis and provides a molecular framework for comparative Evo-Devo studies.


Asunto(s)
Tipificación del Cuerpo/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Esbozos de los Miembros/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Embrión de Pollo , Pollos , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Simulación por Computador , Embrión de Mamíferos , Redes Reguladoras de Genes , Ratones , Proteínas del Tejido Nervioso/metabolismo , RNA-Seq , Especificidad de la Especie , Proteína Gli3 con Dedos de Zinc/metabolismo
6.
Nat Commun ; 12(1): 5557, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34548488

RESUMEN

Precise cis-regulatory control of gene expression is essential for normal embryogenesis and tissue development. The BMP antagonist Gremlin1 (Grem1) is a key node in the signalling system that coordinately controls limb bud development. Here, we use mouse reverse genetics to identify the enhancers in the Grem1 genomic landscape and the underlying cis-regulatory logics that orchestrate the spatio-temporal Grem1 expression dynamics during limb bud development. We establish that transcript levels are controlled in an additive manner while spatial regulation requires synergistic interactions among multiple enhancers. Disrupting these interactions shows that altered spatial regulation rather than reduced Grem1 transcript levels prefigures digit fusions and loss. Two of the enhancers are evolutionary ancient and highly conserved from basal fishes to mammals. Analysing these enhancers from different species reveal the substantial spatial plasticity in Grem1 regulation in tetrapods and basal fishes, which provides insights into the fin-to-limb transition and evolutionary diversification of pentadactyl limbs.


Asunto(s)
Aletas de Animales/metabolismo , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Esbozos de los Miembros/metabolismo , Aletas de Animales/citología , Aletas de Animales/crecimiento & desarrollo , Animales , Secuencia de Bases , Evolución Biológica , Boidae , Bovinos , Pollos , Embrión de Mamíferos , Embrión no Mamífero , Iguanas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Esbozos de los Miembros/citología , Esbozos de los Miembros/crecimiento & desarrollo , Ratones , Ratones Transgénicos , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Conejos , Genética Inversa/métodos , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Tiburones , Transducción de Señal , Porcinos
7.
Elife ; 92020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33006313

RESUMEN

Despite a common understanding that Gli TFs are utilized to convey a Hh morphogen gradient, genetic analyses suggest craniofacial development does not completely fit this paradigm. Using the mouse model (Mus musculus), we demonstrated that rather than being driven by a Hh threshold, robust Gli3 transcriptional activity during skeletal and glossal development required interaction with the basic helix-loop-helix TF Hand2. Not only did genetic and expression data support a co-factorial relationship, but genomic analysis revealed that Gli3 and Hand2 were enriched at regulatory elements for genes essential for mandibular patterning and development. Interestingly, motif analysis at sites co-occupied by Gli3 and Hand2 uncovered mandibular-specific, low-affinity, 'divergent' Gli-binding motifs (dGBMs). Functional validation revealed these dGBMs conveyed synergistic activation of Gli targets essential for mandibular patterning and development. In summary, this work elucidates a novel, sequence-dependent mechanism for Gli transcriptional activity within the craniofacial complex that is independent of a graded Hh signal.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Desarrollo Maxilofacial , Ratones/genética , Proteínas del Tejido Nervioso/genética , Cráneo/crecimiento & desarrollo , Proteína Gli3 con Dedos de Zinc/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Masculino , Desarrollo Maxilofacial/genética , Ratones/metabolismo , Modelos Animales , Proteínas del Tejido Nervioso/metabolismo , Cráneo/metabolismo , Proteína Gli3 con Dedos de Zinc/metabolismo
8.
Curr Top Dev Biol ; 139: 61-88, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32450969

RESUMEN

Vertebrate limb bud outgrowth and patterning is controlled by two instructive signaling centers, the apical ectodermal ridge (AER) and the polarizing region in the posterior limb bud mesenchyme. Molecular analysis of limb bud development has identified a self-regulatory signaling system that operates between the AER and mesenchyme and orchestrates the dynamic progression of limb bud outgrowth and patterning. The first focus of this review are the gene regulatory networks (GRNs) and interactions that control the positioning of the fore- and hindlimb fields along the primary body axis, establish the initial axis polarity and control the precise positioning of the signaling centers. These early processes are largely controlled by activating and inhibiting interactions among types of transcriptional regulators expressed in specific territories. The second focus deals with the dynamic interactions among the GRNs that control limb bud patterning and outgrowth by responding to inputs from the self-regulatory limb bud signaling system. The final part describes the GRN interactions regulating digit morphogenesis and the Turing-type system that controls the periodicity of the digit ray pattern. This review highlights the significant progress made toward an integrative analysis and understanding of the morpho-regulatory systems that orchestrate patterning and outgrowth of vertebrate limb buds in time and space.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Esbozos de los Miembros/metabolismo , Transducción de Señal/genética , Vertebrados/genética , Animales , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Esbozos de los Miembros/embriología , Modelos Genéticos , Vertebrados/clasificación , Vertebrados/embriología
9.
Development ; 146(10)2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31076486

RESUMEN

The key molecular interactions governing vertebrate limb bud development are a paradigm for studying the mechanisms controlling progenitor cell proliferation and specification during vertebrate organogenesis. However, little is known about the cellular heterogeneity of the mesenchymal progenitors in early limb buds that ultimately contribute to the chondrogenic condensations prefiguring the skeleton. We combined flow cytometric and transcriptome analyses to identify the molecular signatures of several distinct mesenchymal progenitor cell populations present in early mouse forelimb buds. In particular, jagged 1 (JAG1)-positive cells located in the posterior-distal mesenchyme were identified as the most immature limb bud mesenchymal progenitors (LMPs), which crucially depend on SHH and FGF signaling in culture. The analysis of gremlin 1 (Grem1)-deficient forelimb buds showed that JAG1-expressing LMPs are protected from apoptosis by GREM1-mediated BMP antagonism. At the same stage, the osteo-chondrogenic progenitors (OCPs) located in the core mesenchyme are already actively responding to BMP signaling. This analysis sheds light on the cellular heterogeneity of the early mouse limb bud mesenchyme and on the distinct response of LMPs and OCPs to morphogen signaling.


Asunto(s)
Proteínas Hedgehog/metabolismo , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Animales , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Hedgehog/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
Open Biol ; 8(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29593116

RESUMEN

The optic fissure is a transient gap in the developing vertebrate eye, which must be closed as development proceeds. A persisting optic fissure, coloboma, is a major cause for blindness in children. Although many genes have been linked to coloboma, the process of optic fissure fusion is still little appreciated, especially on a molecular level. We identified a coloboma in mice with a targeted inactivation of transforming growth factor ß2 (TGFß2). Notably, here the optic fissure margins must have touched, however failed to fuse. Transcriptomic analyses indicated an effect on remodelling of the extracellular matrix (ECM) as an underlying mechanism. TGFß signalling is well known for its effect on ECM remodelling, but it is at the same time often inhibited by bone morphogenetic protein (BMP) signalling. Notably, we also identified two BMP antagonists among the downregulated genes. For further functional analyses we made use of zebrafish, in which we found TGFß ligands expressed in the developing eye, and the ligand binding receptor in the optic fissure margins where we also found active TGFß signalling and, notably, also gremlin 2b (grem2b) and follistatin a (fsta), homologues of the regulated BMP antagonists. We hypothesized that TGFß is locally inducing expression of BMP antagonists within the margins to relieve the inhibition from its regulatory capacity regarding ECM remodelling. We tested our hypothesis and found that induced BMP expression is sufficient to inhibit optic fissure fusion, resulting in coloboma. Our findings can likely be applied also to other fusion processes, especially when TGFß signalling or BMP antagonism is involved, as in fusion processes during orofacial development.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Coloboma/genética , Perfilación de la Expresión Génica/métodos , Factor de Crecimiento Transformador beta2/genética , Animales , Coloboma/tratamiento farmacológico , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Folistatina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Transducción de Señal , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
11.
Cell Rep ; 19(8): 1602-1613, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28538179

RESUMEN

The HAND2 transcriptional regulator controls cardiac development, and we uncover additional essential functions in the endothelial to mesenchymal transition (EMT) underlying cardiac cushion development in the atrioventricular canal (AVC). In Hand2-deficient mouse embryos, the EMT underlying AVC cardiac cushion formation is disrupted, and we combined ChIP-seq of embryonic hearts with transcriptome analysis of wild-type and mutants AVCs to identify the functionally relevant HAND2 target genes. The HAND2 target gene regulatory network (GRN) includes most genes with known functions in EMT processes and AVC cardiac cushion formation. One of these is Snai1, an EMT master regulator whose expression is lost from Hand2-deficient AVCs. Re-expression of Snai1 in mutant AVC explants partially restores this EMT and mesenchymal cell migration. Furthermore, the HAND2-interacting enhancers in the Snai1 genomic landscape are active in embryonic hearts and other Snai1-expressing tissues. These results show that HAND2 directly regulates the molecular cascades initiating AVC cardiac valve development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Redes Reguladoras de Genes , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Animales , Secuencia de Bases , Movimiento Celular/genética , Cromatina/metabolismo , Transición Epitelial-Mesenquimal/genética , Regulación del Desarrollo de la Expresión Génica , Genoma , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Transcripción Genética
12.
Development ; 142(22): 3810-20, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26577204

RESUMEN

The molecular analysis of limb bud development in vertebrates continues to fuel our understanding of the gene regulatory networks that orchestrate the patterning, proliferation and differentiation of embryonic progenitor cells. In recent years, systems biology approaches have moved our understanding of the molecular control of limb organogenesis to the next level by incorporating next generation 'omics' approaches, analyses of chromatin architecture, enhancer-promoter interactions and gene network simulations based on quantitative datasets into experimental analyses. This Review focuses on the insights these studies have given into the gene regulatory networks that govern limb development and into the fin-to-limb transition and digit reductions that occurred during the evolutionary diversification of tetrapod limbs.


Asunto(s)
Evolución Biológica , Tipificación del Cuerpo/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes , Esbozos de los Miembros/embriología , Morfogénesis/fisiología , Transducción de Señal/fisiología , Vertebrados/embriología , Animales , Genes Homeobox/genética , Proteínas Hedgehog/metabolismo , Especificidad de la Especie
13.
PLoS One ; 10(8): e0136566, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26305214

RESUMEN

Studies of mammalian tissue culture cells indicate that the conserved and distinct NDR isoforms, NDR1 and NDR2, play essential cell biological roles. However, mice lacking either Ndr1 or Ndr2 alone develop normally. Here, we studied the physiological consequences of inactivating both NDR1 and NDR2 in mice, showing that the lack of both Ndr1/Ndr2 (called Ndr1/2-double null mutants) causes embryonic lethality. In support of compensatory roles for NDR1 and NDR2, total protein and activating phosphorylation levels of the remaining NDR isoform were elevated in mice lacking either Ndr1 or Ndr2. Mice retaining one single wild-type Ndr allele were viable and fertile. Ndr1/2-double null embryos displayed multiple phenotypes causing a developmental delay from embryonic day E8.5 onwards. While NDR kinases are not required for notochord formation, the somites of Ndr1/2-double null embryos were smaller, irregularly shaped and unevenly spaced along the anterior-posterior axis. Genes implicated in somitogenesis were down-regulated and the normally symmetric expression of Lunatic fringe, a component of the Notch pathway, showed a left-right bias in the last forming somite in 50% of all Ndr1/2-double null embryos. In addition, Ndr1/2-double null embryos developed a heart defect that manifests itself as pericardial edemas, obstructed heart tubes and arrest of cardiac looping. The resulting cardiac insufficiency is the likely cause of the lethality of Ndr1/2-double null embryos around E10. Taken together, we show that NDR kinases compensate for each other in vivo in mouse embryos, explaining why mice deficient for either Ndr1 or Ndr2 are viable. Ndr1/2-double null embryos show defects in somitogenesis and cardiac looping, which reveals their essential functions and shows that the NDR kinases are critically required during the early phase of organogenesis.


Asunto(s)
Organogénesis/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Desarrollo Embrionario/genética , Humanos , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/biosíntesis , Transducción de Señal
14.
PLoS One ; 10(4): e0124870, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25901736

RESUMEN

BACKGROUND: Medulloblastomas are malignant childhood brain tumors that arise due to the aberrant activity of developmental pathways during postnatal cerebellar development and in adult humans. Transcriptome analysis has identified four major medulloblastoma subgroups. One of them, the Sonic hedgehog (SHH) subgroup, is caused by aberrant Hedgehog signal transduction due to mutations in the Patched1 (PTCH1) receptor or downstream effectors. Mice carrying a Patched-1 null allele (Ptch1∆/+) are a good model to study the alterations underlying medulloblastoma development as a consequence of aberrant Hedgehog pathway activity. RESULTS: Transcriptome analysis of human medulloblastomas shows that SERPINE2, also called Protease Nexin-1 (PN-1) is overexpressed in most medulloblastomas, in particular in the SHH and WNT subgroups. As siRNA-mediated lowering of SERPINE2/PN-1 in human medulloblastoma DAOY cells reduces cell proliferation, we analyzed its potential involvement in medulloblastoma development using the Ptch1∆/+ mouse model. In Ptch1∆/+ mice, medulloblastomas arise as a consequence of aberrant Hedgehog pathway activity. Genetic reduction of Serpine2/Pn-1 interferes with medulloblastoma development in Ptch1∆/+ mice, as ~60% of the pre-neoplastic lesions (PNLs) fail to develop into medulloblastomas and remain as small cerebellar nodules. In particular the transcription factor Atoh1, whose expression is essential for development of SHH subgroup medulloblastomas is lost. Comparative molecular analysis reveals the distinct nature of the PNLs in young Ptch1∆/+Pn-1Δ/+ mice. The remaining wild-type Ptch1 allele escapes transcriptional silencing in most cases and the aberrant Hedgehog pathway activity is normalized. Furthermore, cell proliferation and the expression of the cell-cycle regulators Mycn and Cdk6 are significantly reduced in PNLs of Ptch1∆/+Pn-1Δ/+ mice. CONCLUSIONS: Our analysis provides genetic evidence that aberrant Serpine2/Pn-1 is required for proliferation of human and mouse medulloblastoma cells. In summary, our analysis shows that Serpine2/PN-1 boosts malignant progression of PNLs to medulloblastomas, in which the Hedgehog pathway is activated in a SHH ligand-independent manner.


Asunto(s)
Progresión de la Enfermedad , Meduloblastoma/metabolismo , Meduloblastoma/patología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Serpina E2/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Cerebelo/patología , Modelos Animales de Enfermedad , Silenciador del Gen , Genotipo , Proteínas Hedgehog/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/metabolismo , Transducción de Señal
16.
Semin Cell Dev Biol ; 32: 119-27, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24718318

RESUMEN

The analysis of vertebrate limb bud development provides insight of general relevance into the signaling networks that underlie the controlled proliferative expansion of large populations of mesenchymal progenitors, cell fate determination and initiation of differentiation. In particular, extensive genetic analysis of mouse and experimental manipulation of chicken limb bud development has revealed the self-regulatory feedback signaling systems that interlink the main morphoregulatory signaling pathways including BMPs and their antagonists. It this review, we showcase the key role of BMPs and their antagonists during limb bud development. This review provides an understanding of the key morphoregulatory interactions that underlie the highly dynamic changes in BMP activity and signal transduction as limb bud development progresses from initiation and setting-up the signaling centers to determination and formation of the chondrogenic primordia for the limb skeletal elements.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Esbozos de los Miembros/embriología , Esbozos de los Miembros/crecimiento & desarrollo , Transducción de Señal/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Modelos Genéticos , Organogénesis/genética , Organogénesis/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal/genética , Vertebrados
17.
Differentiation ; 85(4-5): 121-30, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23792766

RESUMEN

Endochondral bone development is orchestrated by the spatially and temporally coordinated differentiation of chondrocytes along the longitudinal axis of the cartilage anlage. Initially, the slowly proliferating, periarticular chondrocytes give rise to the pool of rapidly dividing columnar chondrocytes, whose expansion determines the length of the long bones. The Indian hedgehog (IHH) ligand regulates both the proliferation of columnar chondrocytes and their differentiation into post-mitotic hypertrophic chondrocytes in concert with GLI3, one of the main transcriptional effectors of HH signal transduction. In the absence of Hh signalling, the expression of Vlk (vertebrate lonesome kinase, also called Pkdcc) is increased. We now show that the shortening of limb long bones in Vlk-deficient mouse embryos is aggravated by additional inactivation of Gli3. Our analysis establishes that Vlk and Gli3 synergize to control the temporal kinetics of chondrocyte differentiation during long bone development. Whereas differentiation of limb mesenchymal progenitors into chondrocytes and the initial formation of the cartilage anlagen of the limb skeleton are not altered, Vlk and Gli3 are required for the temporally coordinated differentiation of periarticular into columnar and ultimately hypertrophic chondrocytes in long bones. In limbs lacking both Vlk and Gli3, the appearance of columnar and hypertrophic chondrocytes is severely delayed and zones of morphologically distinct chondrocytes are not established until E16.5. At the molecular level, these morphological alterations are reflected by delayed activation and lowered expression of Ihh, Pth1r and Col10a1 in long bone rudiments of double mutant limbs. In summary, our genetic analysis establishes that VLK plays a role in the IHH/GLI3 interactions and that Vlk and Gli3 cooperate to regulate long bone development by modulating the temporal kinetics of establishing columnar and hypertrophic chondrocyte domains.


Asunto(s)
Desarrollo Óseo/genética , Huesos/metabolismo , Diferenciación Celular/genética , Condrocitos/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Proteínas del Tejido Nervioso/genética , Proteínas Quinasas/genética , Animales , Desarrollo Óseo/fisiología , Huesos/citología , Huesos/embriología , Cartílago/metabolismo , Proliferación Celular , Condrocitos/citología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Proteínas Tirosina Quinasas , Transducción de Señal/fisiología , Proteína Gli3 con Dedos de Zinc
18.
BMC Dev Biol ; 12: 23, 2012 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-22888807

RESUMEN

BACKGROUND: Mouse limb bud is a prime model to study the regulatory interactions that control vertebrate organogenesis. Major aspects of limb bud development are controlled by feedback loops that define a self-regulatory signalling system. The SHH/GREM1/AER-FGF feedback loop forms the core of this signalling system that operates between the posterior mesenchymal organiser and the ectodermal signalling centre. The BMP antagonist Gremlin1 (GREM1) is a critical node in this system, whose dynamic expression is controlled by BMP, SHH, and FGF signalling and key to normal progression of limb bud development. Previous analysis identified a distant cis-regulatory landscape within the neighbouring Formin1 (Fmn1) locus that is required for Grem1 expression, reminiscent of the genomic landscapes controlling HoxD and Shh expression in limb buds. RESULTS: Three highly conserved regions (HMCO1-3) were identified within the previously defined critical genomic region and tested for their ability to regulate Grem1 expression in mouse limb buds. Using a combination of BAC and conventional transgenic approaches, a 9 kb region located ~70 kb downstream of the Grem1 transcription unit was identified. This region, termed Grem1 Regulatory Sequence 1 (GRS1), is able to recapitulate major aspects of Grem1 expression, as it drives expression of a LacZ reporter into the posterior and, to a lesser extent, in the distal-anterior mesenchyme. Crossing the GRS1 transgene into embryos with alterations in the SHH and BMP pathways established that GRS1 depends on SHH and is modulated by BMP signalling, i.e. integrates inputs from these pathways. Chromatin immunoprecipitation revealed interaction of endogenous GLI3 proteins with the core cis-regulatory elements in the GRS1 region. As GLI3 is a mediator of SHH signal transduction, these results indicated that SHH directly controls Grem1 expression through the GRS1 region. Finally, all cis-regulatory regions within the Grem1 genomic landscape locate to the DNAse I hypersensitive sites identified in this genomic region by the ENCODE consortium. CONCLUSIONS: This study establishes that distant cis-regulatory regions scattered through a larger genomic landscape control the highly dynamic expression of Grem1, which is key to normal progression of mouse limb bud development.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Proteínas Hedgehog/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Esbozos de los Miembros/embriología , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Secuencia Conservada/genética , Citocinas , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Factores de Transcripción de Tipo Kruppel/metabolismo , Esbozos de los Miembros/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Proteína Gli3 con Dedos de Zinc
19.
Wiley Interdiscip Rev Dev Biol ; 1(6): 803-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23799625

RESUMEN

This review focuses predominantly on the human congenital malformations caused by alterations affecting the morphoregulatory gene networks that control early limb bud patterning and outgrowth. Limb defects are among the most frequent congenital malformations in humans that are caused by genetic mutations or teratogenic effects resulting either in abnormal, loss of, or additional skeletal elements. Spontaneous and engineered mouse models have been used to identify and study the molecular alterations and disrupted gene networks that underlie human congenital limb malformations. More recently, mouse genetics has begun to reveal the alterations that affect the often-large cis-regulatory landscapes that control gene expression in limb buds and cause devastating effects on limb bud development. These findings have paved the way to identifying mutations in cis-regulatory regions as causal to an increasing number of congenital limb malformations in humans. In these cases, no mutations in the coding region of a presumed candidate were previously detected. This review highlights how the current understanding of the molecular gene networks and interactions that control mouse limb bud development provides insight into the etiology of human congenital limb malformations.


Asunto(s)
Deformidades Congénitas de las Extremidades/genética , Animales , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Deformidades Congénitas de las Extremidades/metabolismo , Receptores Patched , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transducción de Señal , Teratógenos/metabolismo
20.
Development ; 138(10): 1913-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21471156

RESUMEN

The essential roles of SHH in anteroposterior (AP) and AER-FGF signalling in proximodistal (PD) limb bud development are well understood. In addition, these morphoregulatory signals are key components of the self-regulatory SHH/GREM1/AER-FGF feedback signalling system that regulates distal progression of limb bud development. This study uncovers an additional signalling module required for coordinated progression of limb bud axis development. Transcriptome analysis using Shh-deficient mouse limb buds revealed that the expression of proximal genes was distally extended from early stages onwards, which pointed to a more prominent involvement of SHH in PD limb axis development. In particular, retinoic acid (RA) target genes were upregulated proximally, while the expression of the RA-inactivating Cyp26b1 enzyme was downregulated distally, pointing to increased RA activity in Shh-deficient mouse limb buds. Further genetic and molecular analysis established that Cyp26b1 expression is regulated by AER-FGF signalling. During initiation of limb bud outgrowth, the activation of Cyp26b1 expression creates a distal 'RA-free' domain, as indicated by complementary downregulation of a transcriptional sensor of RA activity. Subsequently, Cyp26b1 expression increases as a consequence of SHH-dependent upregulation of AER-FGF signalling. To better understand the underlying signalling interactions, computational simulations of the spatiotemporal expression patterns and interactions were generated. These simulations predicted the existence of an antagonistic AER-FGF/CYP26B1/RA signalling module, which was verified experimentally. In summary, SHH promotes distal progression of limb development by enhancing CYP26B1-mediated RA clearance as part of a signalling network linking the SHH/GREM1/AER-FGF feedback loop to the newly identified AER-FGF/CYP26B1/RA module.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Tretinoina/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/genética , Ectodermo/embriología , Ectodermo/metabolismo , Activación Enzimática , Retroalimentación Fisiológica , Femenino , Factores de Crecimiento de Fibroblastos/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Ácido Retinoico 4-Hidroxilasa , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...