Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Dev Dyn ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087648

ABSTRACT

BACKGROUND: Meis family of transcription factors operates in Pbx-Meis-Hox regulatory network controlling development of various tissues including eye, limbs, heart, hindbrain or craniofacial skeletal elements originating from the neural crest. Although studies in mouse provide abundant information about Meis factors function in embryogenesis, little is known about their role in zebrafish. RESULTS: We generated zebrafish lines carrying null mutations in meis1a, meis1b, meis2a, and meis2b genes. Only meis1b mutants are lethal at larval stage around 13 dpf whereas the other mutant lines are viable and fertile. We focused on development of neural crest-derived craniofacial structures such as tendons, cranial nerves, cartilage and accompanying muscles. Meis1b mutants displayed morphogenetic abnormalities in the cartilage originating from the first and second pharyngeal arches. Meckel's cartilage was shorter and wider with fused anterior symphysis and abnormal chondrocyte organization. This resulted in impaired tendons and muscle fiber connections while tenocyte development was not largely affected. CONCLUSIONS: Loss-of-function mutation in meis1b affects cartilage morphology in the lower jaw that leads to disrupted organization of muscles and tendons.

2.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34032267

ABSTRACT

The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.


Subject(s)
Choroid Plexus/embryology , Epithelium/metabolism , Fourth Ventricle/embryology , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Wnt-5a Protein/metabolism , Animals , Brain/embryology , CRISPR-Cas Systems/genetics , Cell Line , Epithelial Cells/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction/physiology , Wnt-5a Protein/genetics
3.
J Biol Chem ; 295(16): 5449-5460, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32169905

ABSTRACT

Haploinsufficiency of Meis homeobox 2 (MEIS2), encoding a transcriptional regulator, is associated with human cleft palate, and Meis2 inactivation leads to abnormal palate development in mice, implicating MEIS2 functions in palate development. However, its functional mechanisms remain unknown. Here we observed widespread MEIS2 expression in the developing palate in mice. Wnt1Cre -mediated Meis2 inactivation in cranial neural crest cells led to a secondary palate cleft. Importantly, about half of the Wnt1Cre ;Meis2f/f mice exhibited a submucous cleft, providing a model for studying palatal bone formation and patterning. Consistent with complete absence of palatal bones, the results from integrative analyses of MEIS2 by ChIP sequencing, RNA-Seq, and an assay for transposase-accessible chromatin sequencing identified key osteogenic genes regulated directly by MEIS2, indicating that it plays a fundamental role in palatal osteogenesis. De novo motif analysis uncovered that the MEIS2-bound regions are highly enriched in binding motifs for several key osteogenic transcription factors, particularly short stature homeobox 2 (SHOX2). Comparative ChIP sequencing analyses revealed genome-wide co-occupancy of MEIS2 and SHOX2 in addition to their colocalization in the developing palate and physical interaction, suggesting that SHOX2 and MEIS2 functionally interact. However, although SHOX2 was required for proper palatal bone formation and was a direct downstream target of MEIS2, Shox2 overexpression failed to rescue the palatal bone defects in a Meis2-mutant background. These results, together with the fact that Meis2 expression is associated with high osteogenic potential and required for chromatin accessibility of osteogenic genes, support a vital function of MEIS2 in setting up a ground state for palatal osteogenesis.


Subject(s)
Homeodomain Proteins/metabolism , Osteogenesis , Palate/metabolism , Animals , Binding Sites , Gene Expression Regulation, Developmental , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Neural Crest/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Palate/embryology , Protein Binding
4.
Int J Mol Sci ; 22(14)2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34299147

ABSTRACT

The mandibular and hyoid arches collectively make up the facial skeleton, also known as the viscerocranium. Although all three germ layers come together to assemble the pharyngeal arches, the majority of tissue within viscerocranial skeletal components differentiates from the neural crest. Since nearly one third of all birth defects in humans affect the craniofacial region, it is important to understand how signalling pathways and transcription factors govern the embryogenesis and skeletogenesis of the viscerocranium. This review focuses on mouse and zebrafish models of craniofacial development. We highlight gene regulatory networks directing the patterning and osteochondrogenesis of the mandibular and hyoid arches that are actually conserved among all gnathostomes. The first part of this review describes the anatomy and development of mandibular and hyoid arches in both species. The second part analyses cell signalling and transcription factors that ensure the specificity of individual structures along the anatomical axes. The third part discusses the genes and molecules that control the formation of bone and cartilage within mandibular and hyoid arches and how dysregulation of molecular signalling influences the development of skeletal components of the viscerocranium. In conclusion, we notice that mandibular malformations in humans and mice often co-occur with hyoid malformations and pinpoint the similar molecular machinery controlling the development of mandibular and hyoid arches.


Subject(s)
Body Patterning , Cartilage/embryology , Hyoid Bone/embryology , Mandible/embryology , Animals , Cartilage/cytology
6.
Dev Biol ; 433(1): 47-60, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29137925

ABSTRACT

Polycomb repressive complexes maintain transcriptional repression of genes encoding crucial developmental regulators through chromatin modification. Here we investigated the role of Polycomb repressive complex 2 (PRC2) in retinal development by inactivating its key components Eed and Ezh2. Conditional deletion of Ezh2 resulted in a partial loss of PRC2 function and accelerated differentiation of Müller glial cells. In contrast, inactivation of Eed led to the ablation of PRC2 function at early postnatal stage. Cell proliferation was reduced and retinal progenitor cells were significantly decreased in this mutant, which subsequently caused depletion of Müller glia, bipolar, and rod photoreceptor cells, primarily generated from postnatal retinal progenitor cells. Interestingly, the proportion of amacrine cells was dramatically increased at postnatal stages in the Eed-deficient retina. In accordance, multiple transcription factors controlling amacrine cell differentiation were upregulated. Furthermore, ChIP-seq analysis showed that these deregulated genes contained bivalent chromatin (H3K27me3+ H3K4me3+). Our results suggest that PRC2 is required for proliferation in order to maintain the retinal progenitor cells at postnatal stages and for retinal differentiation by controlling amacrine cell generation.


Subject(s)
Polycomb Repressive Complex 2/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Histones/metabolism , Methylation , Mice , Neurogenesis , Neuroglia/metabolism , Retina/metabolism , Retina/physiology , Stem Cells/cytology , Stem Cells/metabolism
7.
Development ; 143(12): 2206-16, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27302397

ABSTRACT

The neural crest (NC) is crucial for the evolutionary diversification of vertebrates. NC cells are induced at the neural plate border by the coordinated action of several signaling pathways, including Wnt/ß-catenin. NC cells are normally generated in the posterior neural plate border, whereas the anterior neural fold is devoid of NC cells. Using the mouse model, we show here that active repression of Wnt/ß-catenin signaling is required for maintenance of neuroepithelial identity in the anterior neural fold and for inhibition of NC induction. Conditional inactivation of Tcf7l1, a transcriptional repressor of Wnt target genes, leads to aberrant activation of Wnt/ß-catenin signaling in the anterior neuroectoderm and its conversion into NC. This reduces the developing prosencephalon without affecting the anterior-posterior neural character. Thus, Tcf7l1 defines the border between the NC and the prospective forebrain via restriction of the Wnt/ß-catenin signaling gradient.


Subject(s)
Cell Lineage , Neural Crest/cytology , Transcription Factor 7-Like 1 Protein/metabolism , Zebrafish Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Transdifferentiation , Gene Deletion , Humans , Integrases/metabolism , Mice, Transgenic , Neural Crest/metabolism , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Phenotype , Prosencephalon/embryology , Prosencephalon/metabolism , Repressor Proteins/metabolism , Transcription Factor AP-2/metabolism , Wnt Signaling Pathway , Zebrafish/metabolism , beta Catenin/metabolism
8.
PLoS Genet ; 12(12): e1006441, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27918583

ABSTRACT

Lens induction is a classical developmental model allowing investigation of cell specification, spatiotemporal control of gene expression, as well as how transcription factors are integrated into highly complex gene regulatory networks (GRNs). Pax6 represents a key node in the gene regulatory network governing mammalian lens induction. Meis1 and Meis2 homeoproteins are considered as essential upstream regulators of Pax6 during lens morphogenesis based on their interaction with the ectoderm enhancer (EE) located upstream of Pax6 transcription start site. Despite this generally accepted regulatory pathway, Meis1-, Meis2- and EE-deficient mice have surprisingly mild eye phenotypes at placodal stage of lens development. Here, we show that simultaneous deletion of Meis1 and Meis2 in presumptive lens ectoderm results in arrested lens development in the pre-placodal stage, and neither lens placode nor lens is formed. We found that in the presumptive lens ectoderm of Meis1/Meis2 deficient embryos Pax6 expression is absent. We demonstrate using chromatin immunoprecipitation (ChIP) that in addition to EE, Meis homeoproteins bind to a remote, ultraconserved SIMO enhancer of Pax6. We further show, using in vivo gene reporter analyses, that the lens-specific activity of SIMO enhancer is dependent on the presence of three Meis binding sites, phylogenetically conserved from man to zebrafish. Genetic ablation of EE and SIMO enhancers demostrates their requirement for lens induction and uncovers an apparent redundancy at early stages of lens development. These findings identify a genetic requirement for Meis1 and Meis2 during the early steps of mammalian eye development. Moreover, they reveal an apparent robustness in the gene regulatory mechanism whereby two independent "shadow enhancers" maintain critical levels of a dosage-sensitive gene, Pax6, during lens induction.


Subject(s)
Eye/growth & development , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lens, Crystalline/growth & development , Neoplasm Proteins/genetics , PAX6 Transcription Factor/genetics , Animals , Binding Sites , Ectoderm/growth & development , Ectoderm/pathology , Enhancer Elements, Genetic/genetics , Eye/metabolism , Eye/pathology , Gene Expression Regulation, Developmental , Gene Regulatory Networks/genetics , Humans , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism , PAX6 Transcription Factor/metabolism , Zebrafish/genetics
9.
BMC Dev Biol ; 15: 40, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26545946

ABSTRACT

BACKGROUND: TALE-class homeodomain transcription factors Meis and Pbx play important roles in formation of the embryonic brain, eye, heart, cartilage or hematopoiesis. Loss-of-function studies of Pbx1, 2 and 3 and Meis1 documented specific functions in embryogenesis, however, functional studies of Meis2 in mouse are still missing. We have generated a conditional allele of Meis2 in mice and shown that systemic inactivation of the Meis2 gene results in lethality by the embryonic day 14 that is accompanied with hemorrhaging. RESULTS: We show that neural crest cells express Meis2 and Meis2-defficient embryos display defects in tissues that are derived from the neural crest, such as an abnormal heart outflow tract with the persistent truncus arteriosus and abnormal cranial nerves. The importance of Meis2 for neural crest cells is further confirmed by means of conditional inactivation of Meis2 using crest-specific AP2α-IRES-Cre mouse. Conditional mutants display perturbed development of the craniofacial skeleton with severe anomalies in cranial bones and cartilages, heart and cranial nerve abnormalities. CONCLUSIONS: Meis2-null mice are embryonic lethal. Our results reveal a critical role of Meis2 during cranial and cardiac neural crest cells development in mouse.


Subject(s)
Cranial Nerves/embryology , Heart/embryology , Homeodomain Proteins/genetics , Neural Crest/embryology , Skull/embryology , Animals , Cartilage/abnormalities , Cartilage/embryology , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Hemorrhage/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Crest/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Skull/innervation
10.
Dev Genes Evol ; 225(2): 121-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25689933

ABSTRACT

Wnt/ß-catenin signaling plays an essential role in the retinal pigment epithelium (RPE) determination. Since activity of Pax6 (together with Pax2) is also required for the RPE determination, we investigated a possible genetic interaction between Pax6 and Wnt/ß-catenin signaling pathway by analyzing Pax6, ß-catenin, and Pax6/ß-catenin conditional knockout mice. Although Pax6 inactivation alone had no impact on initial specification determined by the expression of Mitf and Otx2, melanin pigmentation was reduced in the RPE. This suggests that along with Mitf and Otx2, Pax6 is required for the full differentiation of RPE. Reporter gene assays in vitro suggest that hypopigmentation is at least in part due to the direct regulation of genes encoding enzymes involved in melanin synthesis by Pax6, Mitf, and ß-catenin. The RPE of a ß-catenin/Pax6 double mutant was differentiated into the neural retina; however, the tissue was thinner than that of the conditional ß-catenin mutant due to reduced proliferation. Together, our data demonstrate that Pax6 is required for the RPE differentiation by regulating pigmentation and accountable for hyperproliferation in the transdifferentiated RPE. In this context, Pax6 appears to function as a pleiotropic regulator, directing development of ocular tissues in concert with the signaling pathway and, at the same time, regulating expression of structural component of the eye, such as shielding pigment.


Subject(s)
Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Retinal Pigment Epithelium/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Transdifferentiation , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retina/cytology , Retina/metabolism , beta Catenin/genetics
11.
Mol Cell Biochem ; 360(1-2): 289-99, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21935611

ABSTRACT

Tcf3 is a nuclear mediator of canonical Wnt signaling which functions in many systems as a repressor of target gene transcription. In this study, we have cloned and characterized a 6.7 kb fragment of the 5'-flanking promoter region of the mouse Tcf3 gene. In silico analysis of the promoter sequence identified the existence of GC boxes and CpG islands, but failed to identify any TATA box. In addition, the promoter sequence contained putative binding sites for multiple transcription factors, including a few known to regulate the function of mTcf3. Of those, we confirmed functional binding sites for NFκB and Oct1 using a luciferase assay and ChIP. In vitro analysis revealed five potential transcription start sites which resulted in a 298 base pair 5'-untranslated region upstream of the mTcf3 translation start site ATG. Using a luciferase assay, we analyzed the activity of the cloned promoter fragment in embryonically derived neural stem cells. The luciferase activity of a 3.5 kb core promoter fragment (-3243/+211) showed up to 40-fold increased activity compared to the empty vector. Addition of sequences 5' to the 3.5 kb core promoter fragment resulted in a 20-fold drop in luciferase activity, indicating the presence of further upstream repressive elements. In vivo analysis of a 4.5 kb promoter fragment (-4303/+211) driving, the expression of EGFP in mouse embryos highly resembled endogenous expression of mTcf3.


Subject(s)
5' Flanking Region , Basic Helix-Loop-Helix Transcription Factors/genetics , Promoter Regions, Genetic , Animals , Base Sequence , Binding Sites , Cells, Cultured , Cloning, Molecular , Codon, Initiator , Conserved Sequence , Embryo, Mammalian/metabolism , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Molecular Sequence Data , NF-kappa B/genetics , Octamer Transcription Factor-1/genetics
12.
Mol Cell Biochem ; 365(1-2): 53-63, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22270545

ABSTRACT

Tcf3 acts as a transcription factor controlling gene expression in canonical Wnt signaling. In this study we show that mouse Tcf3 represses canonical Wnt signaling in mouse neural stem cells and in human HEK 293 cells. We demonstrate that mouse Tcf3 mediates repression of both moderate and high levels of canonical Wnt signaling, by either competing with other members of the Tcf/Lef family for binding to ß-catenin, or for binding to DNA. We observed that the repressor activity of mouse Tcf3 was only relieved effectively upon simultaneous disruption of both mechanisms. Immunofluorescence of transfected HEK 293 cells showed co-localization of ß-catenin and Tcf3 in the nucleus of cells transfected with full-length Tcf3, but not in cells transfected with N-terminal deleted versions. A direct physical interaction between ß-catenin and Tcf3 in the nucleus was confirmed by co-immunoprecipitation studies. The inhibitory ß-catenin/Tcf3 interface was independent of the ability of Tcf3 to directly interact with DNA.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Regulatory Sequences, Nucleic Acid , Wnt Signaling Pathway , beta Catenin/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Cell Nucleus/metabolism , Cells, Cultured , Conserved Sequence , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Molecular Sequence Data , Neural Stem Cells , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Sequence Deletion , Transcription, Genetic
13.
Front Cell Dev Biol ; 10: 951063, 2022.
Article in English | MEDLINE | ID: mdl-36247013

ABSTRACT

A vertebrate skull is composed of many skeletal elements which display enormous diversity of shapes. Cranial bone formation embodies a multitude of processes, i.e., epithelial-mesenchymal induction, mesenchymal condensation, and endochondral or intramembranous ossification. Molecular pathways determining complex architecture and growth of the cranial skeleton during embryogenesis are poorly understood. Here, we present a model of the hyoid apparatus development in Wnt1-Cre2-induced Meis2 conditional knock-out (cKO) mice. Meis2 cKO embryos develop an aberrant hyoid apparatus-a complete skeletal chain from the base of the neurocranium to lesser horns of the hyoid, resembling extreme human pathologies of the hyoid-larynx region. We examined key stages of hyoid skeletogenesis to obtain a complex image of the hyoid apparatus formation. Lack of Meis2 resulted in ectopic loci of mesenchymal condensations, ectopic cartilage and bone formation, disinhibition of skeletogenesis, and elevated proliferation of cartilage precursors. We presume that all these mechanisms contribute to formation of the aberrant skeletal chain in the hyoid region. Moreover, Meis2 cKO embryos exhibit severely reduced expression of PBX1 and HAND2 in the hyoid region. Altogether, MEIS2 in conjunction with PBX1 and HAND2 affects mesenchymal condensation, specification and proliferation of cartilage precursors to ensure development of the anatomically correct hyoid apparatus.

14.
Cell Mol Neurobiol ; 31(5): 715-27, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21424551

ABSTRACT

Pluripotent embryonic stem cells (ESCs) are able to differentiate into all cell types in the organism including cortical neurons. To follow the dynamic generation of progenitors of the dorsal forebrain in vitro, we generated ESCs from D6-GFP mice in which GFP marks neocortical progenitors and neurons after embryonic day (E) 10.5. We used several cell culture protocols for differentiation of ESCs into progenitors and neurons of the dorsal forebrain. In cell culture, GFP-positive cells were induced under differentiation conditions in quickly formed embryoid bodies (qEBs) after 10-12 day incubation. Activation of Wnt signaling during ESC differentiation further stimulated generation of D6-GFP-positive cortical cells. In contrast, differentiation protocols using normal embryoid bodies (nEBs) yielded only a few D6-GFP-positive cells. Gene expression analysis revealed that multiple components of the canonical Wnt signaling pathway were expressed during the development of embryoid bodies. As shown by immunohistochemistry and quantitative qRT-PCR, D6-GFP-positive cells from qEBs expressed genes that are characteristic for the dorsal forebrain such as Pax6, Dach1, Tbr1, Tbr2, or Sox5. qEBs culture allowed the formation of a D6-GFP positive pseudo-polarized neuroepithelium with the characteristic presence of N-cadherin at the apical pole resembling the structure of the developing neocortex.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neurons/cytology , Prosencephalon/cytology , Animals , Biomarkers/metabolism , Cell Aggregation , Cryoultramicrotomy , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/metabolism , Organ Specificity , Organogenesis , Prosencephalon/metabolism , Signal Transduction , Telencephalon/cytology , Telencephalon/metabolism , Wnt Proteins/metabolism
15.
Genesis ; 48(2): 86-95, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20027618

ABSTRACT

Lens formation in mouse is critically dependent on proper development of the retinal neuroectoderm that is located close beneath the head surface ectoderm. Signaling from the prospective retina triggers lens-specific gene expression in the surface-ectoderm. Supression of canonical Wnt/beta-catenin signaling in the surface ectoderm is one of the prerequisites for lens development because, as we show here, ectopic Wnt activation in the retina and lens abrogates lens formation. Wnt inhibiton is mediated by signals coming from the retina but its exact mechanism is unknown. We show that Pax6 directly controls expression of several Wnt inhibitors such as Sfrp1, Sfrp2, and Dkk1 in the presumptive lens. In accordance, absence of Pax6 function leads to aberrant canonical Wnt activity in the presumptive lens that subsequently impairs lens development. Thus Pax6 is required for down-regulation of canonical Wnt signaling in the presumptive lens ectoderm.


Subject(s)
Ectoderm/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Lens, Crystalline/metabolism , Morphogenesis/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , beta Catenin/metabolism , Animals , Embryo, Mammalian/metabolism , Eye Proteins/genetics , Homeodomain Proteins/genetics , Lens, Crystalline/embryology , Mice , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retina/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
16.
Biol Open ; 9(6)2020 07 02.
Article in English | MEDLINE | ID: mdl-32616504

ABSTRACT

Cranial neural crest cells (cNCCs) originate in the anterior neural tube and populate pharyngeal arches in which they contribute to formation of bone and cartilage. This cell population also provides molecular signals for the development of tissues of non-neural crest origin, such as the tongue muscles, teeth enamel or gland epithelium. Here we show that the transcription factor Meis2 is expressed in the oral region of the first pharyngeal arch (PA1) and later in the tongue primordium. Conditional inactivation of Meis2 in cNCCs resulted in loss of Sonic hedgehog signalling in the oropharyngeal epithelium and impaired patterning of PA1 along the lateral-medial and oral-aboral axis. Failure of molecular specification of PA1, illustrated by altered expression of Hand1/2, Dlx5, Barx1, Gsc and other markers, led to hypoplastic tongue and ectopic ossification of the mandible. Meis2-mutant mice thus display craniofacial defects that are reminiscent of several human syndromes and patients with mutations in the Meis2 gene.


Subject(s)
Body Patterning , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Mandible/embryology , Neural Crest/cytology , Neural Crest/embryology , Signal Transduction , Alleles , Animals , Biomarkers , Body Patterning/genetics , Calcinosis/genetics , Calcinosis/metabolism , Dental Arch/embryology , Gene Deletion , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Organogenesis/genetics , Phenotype
17.
Dev Biol ; 311(1): 223-37, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17916349

ABSTRACT

Neurogenesis in the developing neocortex is a strictly regulated process of cell division and differentiation. Here we report that a gradual retreat of canonical Wnt signaling in the cortex from lateral-to-medial and anterior-to-posterior is a prerequisite of neurogenesis. Ectopic expression of a beta-catenin/LEF1 fusion protein maintains active canonical Wnt signaling in the developing cortex and delays the expression onset of the neurogenic factors Pax6, Ngn2 and Tbr2 and subsequent neurogenesis. Contrary to this, conditional ablation of beta-catenin accelerates expression of the same neurogenic genes. Furthermore, we show that a sustained canonical Wnt activity in the lateral cortex gives rise to cells with hippocampal characteristics in the cortical plate at the expense of the cortical fate, and to cells with dentate gyrus characteristics in the hippocampus. This suggests that the dose of canonical Wnt signaling determines cellular fate in the developing cortex and hippocampus, and that recession of Wnt signaling acts as a morphogenetic gradient regulating neurogenesis in the cortex.


Subject(s)
Hippocampus/cytology , Hippocampus/embryology , Morphogenesis , Signal Transduction , Wnt Proteins/metabolism , Animals , Central Nervous System/embryology , Mice , Mice, Transgenic
18.
Evodevo ; 9: 14, 2018.
Article in English | MEDLINE | ID: mdl-29942461

ABSTRACT

BACKGROUND: Wnt/ß-catenin (or canonical) signalling pathway activity is necessary and used independently several times for specification of vegetal fate and endoderm, gut differentiation, maintenance of epithelium in adult intestine and the development of gut-derived organs in various vertebrate and non-vertebrate organisms. However, its conservation in later stages of digestive tract development still remains questionable due to the lack of detailed data, mainly from Spiralia. RESULTS: Here we characterize the Pdu-Tcf gene, a Tcf/LEF orthologue and a component of Wnt/ß-catenin pathway from Platynereis dumerilii, a spiralian, marine annelid worm. Pdu-Tcf undergoes extensive alternative splicing in the C-terminal region of the gene generating as many as eight mRNA isoforms some of which differ in the presence or absence of a C-clamp domain which suggests a distinct DNA binding activity of individual protein variants. Pdu-Tcf is broadly expressed throughout development which is indicative of many functions. One of the most prominent domains that exhibits rather strong Pdu-Tcf expression is in the putative precursors of endodermal gut cells which are detected after 72 h post-fertilization (hpf). At day 5 post-fertilization (dpf), Pdu-Tcf is expressed in the hindgut and pharynx (foregut), whereas at 7 dpf stage, it is strongly transcribed in the now-cellularized midgut for the first time. In order to gain insight into the role of Wnt/ß-catenin signalling, we disrupted its activity using pharmacological inhibitors between day 5 and 7 of development. The inhibition of Wnt/ß-catenin signalling led to the loss of midgut marker genes Subtilisin-1, Subtilisin-2, α-Amylase and Otx along with a drop in ß-catenin protein levels, Axin expression in the gut and nearly the complete loss of proliferative activity throughout the body of larva. At the same time, a hindgut marker gene Legumain was expanded to the midgut compartment under the same conditions. CONCLUSIONS: Our findings suggest that high Wnt/ß-catenin signalling in the midgut might be necessary for proper differentiation of the endoderm to an epithelium capable of secreting digestive enzymes. Together, our data provide evidence for the role of Wnt/ß-catenin signalling in gut differentiation in Platynereis.

19.
Neural Dev ; 13(1): 8, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29751817

ABSTRACT

Generation of neurons in the embryonic neocortex is a balanced process of proliferation and differentiation of neuronal progenitor cells. Canonical Wnt signalling is crucial for expansion of radial glial cells in the ventricular zone and for differentiation of intermediate progenitors in the subventricular zone. We detected abundant expression of two transcrtiption factors mediating canonical Wnt signalling, Tcf7L1 and Tcf7L2, in the ventricular zone of the embryonic neocortex. Conditional knock-out analysis showed that Tcf7L2, but not Tcf7L1, is the principal Wnt mediator important for maintenance of progenitor cell identity in the ventricular zone. In the absence of Tcf7L2, the Wnt activity is reduced, ventricular zone markers Pax6 and Sox2 are downregulated and the neuroepithelial structure is severed due to the loss of apical adherens junctions. This results in decreased proliferation of radial glial cells, the reduced number of intermediate progenitors in the subventricular zone and hypoplastic forebrain. Our data show that canonical Wnt signalling, which is essential for determining the neuroepithelial character of the neocortical ventricular zone, is mediated by Tcf7L2.


Subject(s)
Neocortex/cytology , Neocortex/embryology , Neurogenesis/physiology , Neurons/physiology , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Cell Count , Cell Differentiation/genetics , Cell Proliferation/genetics , Chloride-Bicarbonate Antiporters , Down-Regulation/genetics , Embryo, Mammalian , Hippocampus/cytology , Hippocampus/embryology , Mice , Mice, Transgenic , Mutation/genetics , Neural Stem Cells/physiology , Neuroglia , Retinal Ganglion Cells/physiology , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics , T-Box Domain Proteins/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Wnt Proteins/metabolism
20.
Mech Dev ; 110(1-2): 179-82, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11744379

ABSTRACT

We have created a transgenic mouse line that expresses Cre recombinase under the control of the novel mouse promoter/enhancer D6. We describe the expression pattern of D6-Cre in a Gtrosa26 reporter background as assayed by LacZ activity. The enhancer activity starts at 10.5 days post-coitum in the telencephalon and is at the later embryonic stages highly restricted to the hippocampus and the neocortex. In adult mice D6-derived cells are found in cortical layers II-VI, in the granular cells of the dentate gyrus and in hippocampal fields CA1-CA3. D6-Cre activity is also detected in the ependymal and subependymal zone of the lateral ventricles which is known to harbor neural stem cells.


Subject(s)
Enhancer Elements, Genetic , Hippocampus/embryology , Hippocampus/enzymology , Integrases/genetics , Neocortex/embryology , Neocortex/enzymology , Viral Proteins/genetics , Animals , Cells, Cultured , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Genes, Reporter , Hippocampus/growth & development , Immunohistochemistry , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Neocortex/growth & development , Neurons/enzymology , Stem Cells/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL