Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38828908

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Gene Expression Regulation, Developmental , Limb Buds , T-Box Domain Proteins , Animals , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Limb Buds/metabolism , Limb Buds/embryology , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Up-Regulation/genetics , Body Patterning/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mesoderm/metabolism , Mesoderm/embryology
2.
STAR Protoc ; 4(4): 102603, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37742180

ABSTRACT

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.


Subject(s)
Embryo, Mammalian , RNA , Animals , Mice , RNA/metabolism , In Situ Hybridization, Fluorescence/methods , Embryo, Mammalian/diagnostic imaging , Embryo, Mammalian/metabolism , Fluorescent Antibody Technique
3.
Nat Commun ; 14(1): 3993, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37414772

ABSTRACT

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.


Subject(s)
Gene Regulatory Networks , Transcription Factors , Animals , Mice , Homeodomain Proteins/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Development ; 148(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34822715

ABSTRACT

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.


Subject(s)
Body Patterning , Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/metabolism , Limb Buds/embryology , Signal Transduction , Smad4 Protein/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hindlimb/embryology , Mice , Mice, Transgenic , Smad4 Protein/genetics
5.
Nat Commun ; 12(1): 5685, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34584102

ABSTRACT

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.


Subject(s)
Body Patterning/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Limb Buds/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chick Embryo , Chickens , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation Sequencing , Computer Simulation , Embryo, Mammalian , Gene Regulatory Networks , Mice , Nerve Tissue Proteins/metabolism , RNA-Seq , Species Specificity , Zinc Finger Protein Gli3/metabolism
6.
Nat Commun ; 12(1): 5557, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548488

ABSTRACT

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.


Subject(s)
Animal Fins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Limb Buds/metabolism , Animal Fins/cytology , Animal Fins/growth & development , Animals , Base Sequence , Biological Evolution , Boidae , Cattle , Chickens , Embryo, Mammalian , Embryo, Nonmammalian , Iguanas , Intercellular Signaling Peptides and Proteins/metabolism , Limb Buds/cytology , Limb Buds/growth & development , Mice , Mice, Transgenic , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rabbits , Reverse Genetics/methods , Sequence Alignment , Sequence Homology, Nucleic Acid , Sharks , Signal Transduction , Swine
7.
Elife ; 92020 10 02.
Article in English | MEDLINE | ID: mdl-33006313

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Maxillofacial Development , Mice/genetics , Nerve Tissue Proteins/genetics , Skull/growth & development , Zinc Finger Protein Gli3/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Male , Maxillofacial Development/genetics , Mice/metabolism , Models, Animal , Nerve Tissue Proteins/metabolism , Skull/metabolism , Zinc Finger Protein Gli3/metabolism
8.
Nat Commun ; 11(1): 2491, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32427842

ABSTRACT

Hox genes encode transcription factors (TFs) that establish morphological diversity in the developing embryo. The similar DNA-binding motifs of the various HOX TFs contrast with the wide-range of HOX-dependent genetic programs. The influence of the chromatin context on HOX binding specificity remains elusive. Here, we used the developing limb as a model system to compare the binding specificity of HOXA13 and HOXD13 (HOX13 hereafter), which are required for digit formation, and HOXA11, involved in forearm/leg development. We find that upon ectopic expression in distal limb buds, HOXA11 binds sites normally HOX13-specific. Importantly, these sites are loci whose chromatin accessibility relies on HOX13. Moreover, we show that chromatin accessibility specific to the distal limb requires HOX13 function. Based on these results, we propose that HOX13 TFs pioneer the distal limb-specific chromatin accessibility landscape for the proper implementation of the distal limb developmental program.


Subject(s)
Chromatin/genetics , Forelimb/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Limb Buds/metabolism , Animals , Binding Sites/genetics , Chromatin/metabolism , Forelimb/embryology , Gene Expression Profiling/methods , Homeodomain Proteins/metabolism , Limb Buds/embryology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding
9.
Curr Top Dev Biol ; 139: 61-88, 2020.
Article in English | MEDLINE | ID: mdl-32450969

ABSTRACT

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.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Limb Buds/metabolism , Signal Transduction/genetics , Vertebrates/genetics , Animals , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Limb Buds/embryology , Models, Genetic , Vertebrates/classification , Vertebrates/embryology
10.
Cell Rep ; 31(1): 107490, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268095

ABSTRACT

Digit loss/reductions are evolutionary adaptations in cursorial mammals such as pigs. To gain mechanistic insight into these processes, we performed a comparative molecular analysis of limb development in mouse and pig embryos, which revealed a loss of anterior-posterior polarity during distal progression of pig limb bud development. These alterations in pig limb buds are paralleled by changes in the mesenchymal response to Sonic hedgehog (SHH) signaling, which is altered upstream of the reduction and loss of Fgf8 expression in the ectoderm that overlaps the reduced and vestigial digit rudiments of the pig handplate, respectively. Furthermore, genome-wide open chromatin profiling using equivalent developmental stages of mouse and pig limb buds reveals the functional divergence of about one-third of the regulatory genome. This study uncovers widespread alterations in the regulatory landscapes of genes essential for limb development that likely contributed to the morphological diversion of artiodactyl limbs from the pentadactyl archetype of tetrapods.


Subject(s)
Body Patterning/genetics , Limb Buds/embryology , Limb Buds/metabolism , Animals , Biological Evolution , Ectoderm/metabolism , Extremities/embryology , Female , Gene Expression Regulation, Developmental/genetics , Male , Mesoderm/metabolism , Mice/embryology , Phenotype , Polydactyly/genetics , Signal Transduction/genetics , Swine/embryology , Trans-Activators/metabolism
11.
Development ; 146(10)2019 05 28.
Article in English | MEDLINE | ID: mdl-31076486

ABSTRACT

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.


Subject(s)
Hedgehog Proteins/metabolism , Limb Buds/embryology , Limb Buds/metabolism , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Signal Transduction/genetics , Signal Transduction/physiology
12.
Stem Cell Reports ; 9(4): 1124-1138, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28919259

ABSTRACT

Bone-derived mesenchymal stromal cells (MSCs) differentiate into multiple lineages including chondro- and osteogenic fates and function in establishing the hematopoietic compartment of the bone marrow. Here, we analyze the emergence of different MSC types during mouse limb and long bone development. In particular, PDGFRαposSCA-1pos (PαS) cells and mouse skeletal stem cells (mSSCs) are detected within the PDGFRαposCD51pos (PαCD51) mesenchymal progenitors, which are the most abundant progenitors in early limb buds and developing long bones until birth. Long-bone-derived PαS cells and mSSCs are most prevalent in newborn mice, and molecular analysis shows that they constitute distinct progenitor populations from the earliest stages onward. Differential expression of CD90 and CD73 identifies four PαS subpopulations that display distinct chondro- and osteogenic differentiation potentials. Finally, we show that cartilage constructs generated from CD90pos PαS cells are remodeled into bone organoids encompassing functional endothelial and hematopoietic compartments, which makes these cells suited for bone tissue engineering.


Subject(s)
Bone Development , Cell Differentiation , Mesenchymal Stem Cells/cytology , Osteogenesis , Animals , Antigens, CD/metabolism , Biomarkers , Cell Lineage , Chondrogenesis , Hematopoiesis , Immunophenotyping , Mesenchymal Stem Cells/metabolism , Mice , Neovascularization, Physiologic , Phenotype
13.
Cell Rep ; 19(8): 1602-1613, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28538179

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Gene Regulatory Networks , Heart Valves/embryology , Heart Valves/metabolism , Animals , Base Sequence , Cell Movement/genetics , Chromatin/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Genome , Mesoderm/cytology , Mesoderm/metabolism , Mice , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transcription, Genetic
14.
J Mech Behav Biomed Mater ; 62: 384-398, 2016 09.
Article in English | MEDLINE | ID: mdl-27258932

ABSTRACT

Ti-Zr alloys have recently started to receive a considerable amount of attention as promising materials for dental applications. This work compares mechanical properties of a new Ti-15Zr alloy to those of commercially pure titanium Grade4 in two surface conditions - machined and modified by sand-blasting and etching (SLA). As a result of significantly smaller grain size in the initial condition (1-2µm), the strength of Ti-15Zr alloy was found to be 10-15% higher than that of Grade4 titanium without reduction in the tensile elongation or compromising the fracture toughness. The fatigue endurance limit of the alloy was increased by around 30% (560MPa vs. 435MPa and 500MPa vs. 380MPa for machined and SLA-treated surfaces, respectively). Additional implant fatigue tests showed enhanced fatigue performance of Ti-15Zr over Ti-Grade4.


Subject(s)
Dental Alloys , Dental Implants , Titanium , Zirconium , Materials Testing , Surface Properties
15.
PLoS One ; 10(8): e0136566, 2015.
Article in English | MEDLINE | ID: mdl-26305214

ABSTRACT

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.


Subject(s)
Organogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Embryonic Development/genetics , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/biosynthesis , Signal Transduction
16.
PLoS One ; 10(4): e0124870, 2015.
Article in English | MEDLINE | ID: mdl-25901736

ABSTRACT

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.


Subject(s)
Disease Progression , Medulloblastoma/metabolism , Medulloblastoma/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Serpin E2/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cerebellum/pathology , Disease Models, Animal , Gene Silencing , Genotype , Hedgehog Proteins/metabolism , Humans , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/metabolism , Signal Transduction
17.
Br J Pharmacol ; 172(10): 2427-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25899710

ABSTRACT

This article discusses the background to the need for change in the reporting of experiments involving animals, including a report of a consensus meeting organised by the Basel Declaration Society and Understanding Animal Research UK that sought to Internationalise guidelines for reporting experiments involving animals. A commentary on the evolution of BJP's attempts to implement the ARRIVE guidelines and details of our new guidance for authors is published separately (McGrath, 2014). This is one of a series of editorials discussing updates to the BJP Instructions to Authors LINKED EDITORIALS: This Editorial is the first in a series. The other Editorials in this series will be published in the forthcoming issues. To view them, visit: http://onlinelibrary.wiley.com/journal/10.1111/(ISSN)1476-5381.


Subject(s)
Animal Experimentation/standards , Disclosure , Editorial Policies , Periodicals as Topic/standards , Research Report/standards , Animals , Humans , Switzerland
18.
Cell Rep ; 9(6): 2071-83, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25497097

ABSTRACT

The basic-helix-loop-helix (bHLH) transcription factor Hand2 plays critical roles during cardiac morphogenesis via expression and function within myocardial, neural crest, and epicardial cell populations. Here, we show that Hand2 plays two essential Notch-dependent roles within the endocardium. Endocardial ablation of Hand2 results in failure to develop a patent tricuspid valve, intraventricular septum defects, and hypotrabeculated ventricles, which collectively resemble the human congenital defect tricuspid atresia. We show endocardial Hand2 to be an integral downstream component of a Notch endocardium-to-myocardium signaling pathway and a direct transcriptional regulator of Neuregulin1. Additionally, Hand2 participates in endocardium-to-endocardium-based cell signaling, with Hand2 mutant hearts displaying an increased density of coronary lumens. Molecular analyses further reveal dysregulation of several crucial components of Vegf signaling, including VegfA, VegfR2, Nrp1, and VegfR3. Thus, Hand2 functions as a crucial downstream transcriptional effector of endocardial Notch signaling during both cardiogenesis and coronary vasculogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endocardium/metabolism , Gene Expression Regulation, Developmental , Receptors, Notch/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Endocardium/embryology , Mice , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Receptors, Notch/genetics , Transcriptional Activation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
19.
Dev Cell ; 31(3): 345-357, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25453830

ABSTRACT

The genetic networks that govern vertebrate development are well studied, but how the interactions of trans-acting factors with cis-regulatory modules (CRMs) are integrated into spatiotemporal regulation of gene expression is not clear. The transcriptional regulator HAND2 is required during limb, heart, and branchial arch development. Here, we identify the genomic regions enriched in HAND2 chromatin complexes from mouse embryos and limb buds. Then we analyze the HAND2 target CRMs in the genomic landscapes encoding transcriptional regulators required in early limb buds. HAND2 controls the expression of genes functioning in the proximal limb bud and orchestrates the establishment of anterior and posterior polarity of the nascent limb bud mesenchyme by impacting Gli3 and Tbx3 expression. TBX3 is required downstream of HAND2 to refine the posterior Gli3 expression boundary. Our analysis uncovers the transcriptional circuits that function in establishing distinct mesenchymal compartments downstream of HAND2 and upstream of SHH signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Extremities/embryology , Gene Expression Regulation, Developmental/physiology , Limb Buds/metabolism , Mesoderm/metabolism , Animals , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL