Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Development ; 149(9)2022 05 01.
Article in English | MEDLINE | ID: mdl-35420656

ABSTRACT

For left-right symmetry breaking in the mouse embryo, the basal body must become positioned at the posterior side of node cells, but the precise mechanism for this has remained unknown. Here, we examined the role of microtubules (MTs) and actomyosin in this basal body positioning. Exposure of mouse embryos to agents that stabilize or destabilize MTs or F-actin impaired such positioning. Active myosin II was detected at the anterior side of node cells before the posterior shift of the basal body, and this asymmetric activation was lost in Prickle and dachsous mutant embryos. The organization of basal-body associated MTs (baMTs) was asymmetric between the anterior and posterior sides of node cells, with anterior baMTs extending horizontally and posterior baMTs extending vertically. This asymmetry became evident after polarization of the PCP core protein Vangl1 and before the posterior positioning of the basal body, and it also required the PCP core proteins Prickle and dachsous. Our results suggest that the asymmetry in baMT organization may play a role in correct positioning of the basal body for left-right symmetry breaking.


Subject(s)
Basal Bodies , Cell Polarity , Actins/metabolism , Animals , Cell Polarity/physiology , Cilia/metabolism , Mice , Microtubules/metabolism
2.
PLoS Genet ; 16(12): e1009232, 2020 12.
Article in English | MEDLINE | ID: mdl-33347437

ABSTRACT

Motile cilia can beat with distinct patterns, but how motility variations are regulated remain obscure. Here, we have studied the role of the coiled-coil protein CFAP53 in the motility of different cilia-types in the mouse. While node (9+0) cilia of Cfap53 mutants were immotile, tracheal and ependymal (9+2) cilia retained motility, albeit with an altered beat pattern. In node cilia, CFAP53 mainly localized at the base (centriolar satellites), whereas it was also present along the entire axoneme in tracheal cilia. CFAP53 associated tightly with microtubules and interacted with axonemal dyneins and TTC25, a dynein docking complex component. TTC25 and outer dynein arms (ODAs) were lost from node cilia, but were largely maintained in tracheal cilia of Cfap53-/- mice. Thus, CFAP53 at the base of node cilia facilitates axonemal transport of TTC25 and dyneins, while axonemal CFAP53 in 9+2 cilia stabilizes dynein binding to microtubules. Our study establishes how differential localization and function of CFAP53 contributes to the unique motion patterns of two important mammalian cilia-types.


Subject(s)
Axonemal Dyneins/metabolism , Axoneme/metabolism , Biological Transport, Active/genetics , Cell Movement/genetics , Cilia/metabolism , Embryo, Mammalian/metabolism , Microtubules/metabolism , Animals , Axonemal Dyneins/genetics , Axoneme/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cilia/genetics , Embryo, Mammalian/physiology , Embryo, Mammalian/ultrastructure , Ependyma/embryology , Ependyma/metabolism , Ependyma/physiology , Fluorescent Antibody Technique , Genotype , Immunoprecipitation , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microtubules/genetics , Mutation , Phenotype , Trachea/embryology , Trachea/metabolism , Trachea/physiology , Trachea/ultrastructure
3.
Am J Hum Genet ; 99(2): 460-9, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27486780

ABSTRACT

Multiprotein complexes referred to as outer dynein arms (ODAs) develop the main mechanical force to generate the ciliary and flagellar beat. ODA defects are the most common cause of primary ciliary dyskinesia (PCD), a congenital disorder of ciliary beating, characterized by recurrent infections of the upper and lower airways, as well as by progressive lung failure and randomization of left-right body asymmetry. Using a whole-exome sequencing approach, we identified recessive loss-of-function mutations within TTC25 in three individuals from two unrelated families affected by PCD. Mice generated by CRISPR/Cas9 technology and carrying a deletion of exons 2 and 3 in Ttc25 presented with laterality defects. Consistently, we observed immotile nodal cilia and missing leftward flow via particle image velocimetry. Furthermore, transmission electron microscopy (TEM) analysis in TTC25-deficient mice revealed an absence of ODAs. Consistent with our findings in mice, we were able to show loss of the ciliary ODAs in humans via TEM and immunofluorescence (IF) analyses. Additionally, IF analyses revealed an absence of the ODA docking complex (ODA-DC), along with its known components CCDC114, CCDC151, and ARMC4. Co-immunoprecipitation revealed interaction between the ODA-DC component CCDC114 and TTC25. Thus, here we report TTC25 as a new member of the ODA-DC machinery in humans and mice.


Subject(s)
Axoneme/genetics , Axoneme/metabolism , Carrier Proteins/genetics , Cilia/pathology , Dyneins/chemistry , Dyneins/metabolism , Kartagener Syndrome/genetics , Kartagener Syndrome/pathology , Mutation , Animals , Axoneme/pathology , Axoneme/ultrastructure , Cilia/metabolism , Cilia/ultrastructure , Dyneins/genetics , Dyneins/ultrastructure , Exome/genetics , Exons/genetics , Fluorescent Antibody Technique , Genes, Recessive , Humans , Mice , Microscopy, Electron, Transmission , Protein Binding , Xenopus , Xenopus Proteins/deficiency , Xenopus Proteins/genetics
4.
Semin Cell Dev Biol ; 32: 80-4, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24704359

ABSTRACT

Two TGFß-related proteins, Nodal and Lefty, are asymmetrically expressed and play central roles in establishing left-right (L-R) asymmetry of our body. Nodal acts as a left-side determinant whereas Lefty restricts Nodal activity to the left side by acting as a feedback inhibitor of Nodal. While the mechanism for symmetry breaking is variable among animals, the pair of Nodal and Lefty has a conserved role in the L-R asymmetry pathway. Function and regulation of Nodal and Lefty have been revealed in the last decades, but in this review we summarize the role of TGFß-related proteins together with more recent findings. We mainly discuss observations made with mouse embryos, unless indicated otherwise.


Subject(s)
Body Patterning/genetics , Left-Right Determination Factors/genetics , Nodal Protein/genetics , Signal Transduction/genetics , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Left-Right Determination Factors/metabolism , Mice , Models, Genetic , Nodal Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
Dev Biol ; 395(2): 331-41, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25224222

ABSTRACT

The transcription factor Pitx2c is expressed in primordial visceral organs in a left-right (L-R) asymmetric manner and executes situs-specific morphogenesis. Here we show that Pitx2c is also L-R asymmetrically expressed in the developing mouse limb. Human PITX2c exhibits the same transcriptional activity in the mouse limb. The asymmetric expression of Pitx2c in the limb also exhibits dorsal-ventral and anterior-posterior polarities, being confined to the posterior-dorsal region of the left limb. Left-sided Pitx2c expression in the limb is regulated by Nodal signaling through a Nodal-responsive enhancer. Pitx2c is expressed in lateral plate mesoderm (LPM)-derived cells in the left limb that contribute to various limb connective tissues. The number of Pitx2c(+) cells in the left limb was found to be negatively regulated by Pitx2c itself. Although obvious defects were not apparent in the limb of mice lacking asymmetric Pitx2c expression, Pitx2c may regulate functional L-R asymmetry of the limb.


Subject(s)
Extremities/embryology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Morphogenesis/physiology , Transcription Factors/metabolism , Animals , DNA Primers/genetics , Fluorescent Antibody Technique , Galactosides , Gene Knock-In Techniques , In Situ Hybridization , Indoles , Mice , Mice, Transgenic , Tamoxifen , Homeobox Protein PITX2
6.
Nature ; 460(7252): 287-91, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19483677

ABSTRACT

Diverse histone modifications are catalysed and recognized by various specific proteins, establishing unique modification patterns that act as transcription signals. In particular, histone H3 trimethylation at lysine 36 (H3K36me3) is associated with actively transcribed regions and has been proposed to provide landmarks for continuing transcription; however, the control mechanisms and functions of H3K36me3 in higher eukaryotes are unknown. Here we show that the H3K36me3-specific histone methyltransferase (HMTase) Wolf-Hirschhorn syndrome candidate 1 (WHSC1, also known as NSD2 or MMSET) functions in transcriptional regulation together with developmental transcription factors whose defects overlap with the human disease Wolf-Hirschhorn syndrome (WHS). We found that mouse Whsc1, one of five putative Set2 homologues, governed H3K36me3 along euchromatin by associating with the cell-type-specific transcription factors Sall1, Sall4 and Nanog in embryonic stem cells, and Nkx2-5 in embryonic hearts, regulating the expression of their target genes. Whsc1-deficient mice showed growth retardation and various WHS-like midline defects, including congenital cardiovascular anomalies. The effects of Whsc1 haploinsufficiency were increased in Nkx2-5 heterozygous mutant hearts, indicating their functional link. We propose that WHSC1 functions together with developmental transcription factors to prevent the inappropriate transcription that can lead to various pathophysiologies.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Wolf-Hirschhorn Syndrome/metabolism , Animals , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Lysine/metabolism , Methylation , Mice , Mice, Inbred C57BL , Nanog Homeobox Protein , Protein Binding , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic
7.
Dev Biol ; 376(1): 23-30, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23357539

ABSTRACT

In the node of mouse embryo, rotational movements of cilia generate an external liquid flow known as nodal flow, which determines left-right asymmetric gene expression. How nodal flow is converted into asymmetric gene expression is still controversial, but the increase of Ca(2+) levels in endodermal cells to the left of the node has been proposed to play a role. However, Ca(2+) signals inside the node itself have not yet been described. By our optimized Ca(2+) imaging method, we were able to observe dynamic Ca(2+) signals in the node in live mouse embryos. Pharmacological disruption of Ca(2+) signals did not affect ciliary movements or nodal flow, but did alter the expression patterns of the Nodal and Cerl-2 genes. Quantitative analyses of Ca(2+) signal frequencies and distributions showed that during left-right axis establishment, formerly symmetric Ca(2+) signals became biased to the left side. In iv/iv mutant embryos that showed randomized laterality due to ciliary immotility, Ca(2+) signals were found to be variously left-sided, right-sided, or bilateral, and thus symmetric on average. In Pkd2 mutant embryos, which lacked polycystin-2, a Ca(2+)-permeable cation channel necessary for left-right axis formation, the Ca(2+) signal frequency was lower than in wild-type embryos. Our data support a model in which dynamic Ca(2+) signals in the node are involved in left-right patterning.


Subject(s)
Body Patterning/physiology , Calcium Signaling/physiology , Gene Expression Regulation, Developmental/physiology , Organizers, Embryonic/embryology , Animals , Cilia/physiology , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Nodal Protein/metabolism , Organizers, Embryonic/metabolism , TRPP Cation Channels/genetics
8.
Dev Biol ; 381(1): 203-12, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23742838

ABSTRACT

Qilin is one of several genes in zebrafish whose mutation results in cystic kidney. We have now studied the role of its mouse ortholog, Cluap1, in embryonic development by generating Cluap1 knockout (Cluap1-/-) mice. Cluap1-/- embryos died mid-gestation manifesting impairment of ciliogenesis in various regions including the node and neural tube. The basal body was found to be properly docked to the apical membrane of cells in the mutant, but the axoneme failed to grow. Cluap1 is a ciliary protein and is preferentially localized at the base and tip of cilia. Hedgehog signaling, as revealed with a Pacthed1-lacZ reporter gene, was lost in Cluap1-/- embryos at embryonic day (E) 8.5 but was ectopically expanded at E9.0. The Cluap1 knockout embryos also failed to manifest left-right asymmetric expression of Nodal in the lateral plate, most likely as a result of the loss of Hedgehog signaling in node crown cells that in turn leads to pronounced down-regulation of Gdf1 expression in these cells. Crown cell-specific restoration of Cluap1 expression rescued Gdf1 expression in crown cells and left-sided Nodal expression in the lateral plate of mutant embryos. Our results suggest that Cluap1 contributes to ciliogenesis by regulating the intraflagellar transport (IFT) cycle at the base and tip of the cilium.


Subject(s)
Cilia/metabolism , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/physiology , Morphogenesis/genetics , Animals , Body Patterning , Down-Regulation , Fibroblasts/metabolism , Genes, Reporter , Genotype , Hedgehog Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lac Operon , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Signal Transduction
9.
Development ; 138(10): 1913-23, 2011 May.
Article in English | MEDLINE | ID: mdl-21471156

ABSTRACT

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.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/metabolism , Limb Buds/embryology , Limb Buds/metabolism , Tretinoin/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Ectoderm/embryology , Ectoderm/metabolism , Enzyme Activation , Feedback, Physiological , Female , Fibroblast Growth Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Mutation , Oligonucleotide Array Sequence Analysis , Pregnancy , Retinoic Acid 4-Hydroxylase , Signal Transduction
10.
Mol Cell Neurosci ; 52: 128-39, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23147109

ABSTRACT

Transcriptional regulation of gene expression during development is critical for proper neuronal differentiation and migration. Alternative splicing and differential isoform expression have been demonstrated for most mammalian genes, but their specific contributions to gene function are not well understood. In mice, the transcription factor gene Pitx2 is expressed as three different isoforms (PITX2A, PITX2B, and PITX2C) which have unique amino termini and common DNA binding homeodomains and carboxyl termini. The specific roles of these isoforms in neuronal development are not known. Here we report the onset of Pitx2ab and Pitx2c isoform-specific expression by E9.5 in the developing mouse brain. Using isoform-specific Pitx2 deletion mouse strains, we show that collicular neuron migration requires PITX2AB and that collicular GABAergic differentiation and targeting of hypothalamic projections require unique Pitx2 isoform dosage. These results provide insights into Pitx2 dosage and isoform-specific requirements underlying midbrain and hypothalamic development.


Subject(s)
Homeodomain Proteins/metabolism , Hypothalamus/embryology , Neurogenesis/physiology , Neurons/metabolism , Superior Colliculi/embryology , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Neurons/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Real-Time Polymerase Chain Reaction , Superior Colliculi/metabolism , Transcription Factors/genetics , Homeobox Protein PITX2
11.
Dev Biol ; 371(2): 136-45, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22819673

ABSTRACT

Tight junctions (TJs) connect epithelial cells and form a semipermeable barrier that only allows selective passage of ions and solutes across epithelia. Here we show that mice lacking EpCAM, a putative cell adhesion protein frequently overexpressed in human cancers, manifest intestinal barrier defects and die shortly after birth as a result of intestinal erosion. EpCAM was found to be highly expressed in the developing intestinal epithelium of wild-type mice and to localize to cell-cell junctions including TJs. Claudin-7 colocalized with EpCAM at cell-cell junctions, and the two proteins were found to associate with each other. Claudins 2, 3, 7, and 15 were down-regulated in the intestine of EpCAM mutant mice, with claudin-7 being reduced to undetectable levels. TJs in the mutant intestinal epithelium were morphologically abnormal with the network of TJ strands scattered and dispersed. Finally, the barrier function of the intestinal epithelium was impaired in the mutant animals. These results suggest that EpCAM contributes to formation of intestinal barrier by recruiting claudins to cell-cell junctions.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Claudins/metabolism , Intestinal Mucosa/metabolism , Tight Junctions/metabolism , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Claudin-3/genetics , Claudin-3/metabolism , Claudins/genetics , Down-Regulation , Epithelial Cell Adhesion Molecule , Intestinal Mucosa/embryology , Mice , Mice, Knockout
12.
Nature ; 450(7167): 285-8, 2007 Nov 08.
Article in English | MEDLINE | ID: mdl-17994097

ABSTRACT

Laterality of the internal organs of vertebrates is determined by asymmetric Nodal signalling in the lateral plate mesoderm. A deficiency of such signalling results in heterotaxia syndrome, characterized by anomalous laterality of visceral organs and complex congenital heart conditions. Pitx2, the transcription factor induced by the Nodal signal, regulates left-right asymmetric morphogenesis. The cellular and molecular bases of asymmetric morphogenesis remain largely unknown, however. Here we show that ablation of unilateral Pitx2 expression in mice impairs asymmetric remodelling of the branchial arch artery (BAA) system, resulting in randomized laterality of the aortic arch. Pitx2-positive cells were found not to contribute to asymmetrically remodelled arteries. Instead, Pitx2 functions in the secondary heart field and induces a dynamic morphological change in the outflow tract of the heart, which results in the provision of an asymmetric blood supply to the sixth BAA. This uneven distribution of blood flow results in differential signalling by both the platelet-derived growth factor receptor and vascular endothelial growth factor receptor 2. The consequent stabilization of the left sixth BAA and regression of its right counterpart underlie left-sided formation of the aortic arch. Our results therefore indicate that haemodynamics, generated by a Pitx2-induced morphological change in the outflow tract, is responsible for the asymmetric remodelling of the great arteries.


Subject(s)
Aorta, Thoracic/embryology , Aorta, Thoracic/metabolism , Hemodynamics/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Morphogenesis , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Aorta, Thoracic/anatomy & histology , Aorta, Thoracic/physiology , Mice , Platelet-Derived Growth Factor/metabolism , Regional Blood Flow , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Homeobox Protein PITX2
13.
Dev Biol ; 353(2): 321-30, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21419113

ABSTRACT

Left-right (L-R) asymmetry in the mouse embryo is generated in the node and is dependent on cilia-driven fluid flow, but how the initial asymmetry is transmitted from the node to the lateral plate has remained unknown. We have now identified a transcriptional enhancer (ANE) in the human LEFTY1 gene that exhibits marked L>R asymmetric activity in perinodal cells of the mouse embryo. Dissection of ANE revealed that it is activated in the perinodal cells on the left side by Nodal signaling, suggesting that Nodal activity in the node is asymmetric at a time when Nodal expression is symmetric. Phosphorylated Smad2/3 (pSmad2) indeed manifested an L-R asymmetric distribution at the node, being detected in perinodal cells preferentially on the left side. This asymmetry in pSmad2 distribution was found to be generated not by unidirectional transport of Nodal but rather as a result of LR distribution of active Nodal in the node is translated into the asymmetry in LPM.


Subject(s)
Body Patterning/genetics , Body Patterning/physiology , Mesoderm/embryology , Nodal Protein/genetics , Nodal Protein/physiology , Animals , Biological Transport, Active , Enhancer Elements, Genetic , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Left-Right Determination Factors/genetics , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Knockout , Mice, Neurologic Mutants , Mice, Transgenic , Phosphorylation , Pregnancy , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
14.
Hum Mol Genet ; 19(7): 1286-301, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20071345

ABSTRACT

Total anomalous pulmonary venous return (TAPVR) is a congenital heart defect inherited via complex genetic and/or environmental factors. We report detailed mapping in extended TAPVR kindreds and mutation analysis in TAPVR patients that implicate the PDGFRA gene in the development of TAPVR. Gene expression studies in mouse and chick embryos for both the Pdgfra receptor and its ligand Pdgf-a show temporal and spatial patterns consistent with a role in pulmonary vein (PV) development. We used an in ovo function blocking assay in chick and a conditional knockout approach in mouse to knock down Pdgfra expression in the developing venous pole during the period of PV formation. We observed that loss of PDGFRA function in both organisms causes TAPVR with low penetrance (approximately 7%) reminiscent of that observed in our human TAPVR kindreds. Intermediate inflow tract anomalies occurred in a higher percentage of embryos (approximately 30%), suggesting that TAPVR occurs at one end of a spectrum of defects. We show that the anomalous pulmonary venous connection seen in chick and mouse is highly similar to TAPVR discovered in an abnormal early stage embryo from the Kyoto human embryo collection. Whereas the embryology of the normal venous pole and PV is becoming understood, little is known about the embryogenesis or molecular pathogenesis of TAPVR. These models of TAPVR provide important insight into the pathogenesis of PV defects. Taken together, these data from human genetics and animal models support a role for PDGF-signaling in normal PV development, and in the pathogenesis of TAPVR.


Subject(s)
Heart Defects, Congenital/genetics , Pulmonary Veins/abnormalities , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , Chick Embryo , Humans , Mice , Mice, Mutant Strains , Models, Animal , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
15.
Dev Dyn ; 240(1): 195-203, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21089073

ABSTRACT

To aid in detection and tracking of cells targeted by the left-right (LR) pathway in the heart throughout morphogenesis, expression from a Pitx2c-lacZ transgene (P2Ztg) was analysed in detail. ß-galactosidase expression from P2Ztg was robust, allowing reliable visualisation of low-level Pitx2c expression, and was virtually entirely dependent upon NODAL signalling in the heart. P2Ztg showed expression in trabecular and septal, as well as non-trabecular, myocardium, and a strong expression bias in myocardium associated with individual endocardial cushions of the atrioventricular canal and outflow tract, which are essential for cardiac septation. Expression on the ventral surface of the outflow tract evolved to a specific stripe that could be used to track the future aorta during outflow tract spiralling and remodelling. Our data show that the P2Ztg transgene is a useful resource for detection of molecular disturbances in the LR cascade, as well as morphogenetic defects associated with other cardiac congenital disorders.


Subject(s)
Genes, Reporter , Homeodomain Proteins/genetics , Myocardium/metabolism , Transcription Factors/genetics , Transgenes , Animals , Gene Expression Regulation, Developmental , Genes, Reporter/physiology , Heart/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Homeodomain Proteins/metabolism , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Morphogenesis/genetics , Neural Crest/metabolism , Reproducibility of Results , Transcription Factors/metabolism , Transgenes/genetics , Transgenes/physiology , Homeobox Protein PITX2
16.
Dev Cell ; 10(4): 451-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16580991

ABSTRACT

The earliest recognizable sign of patterning of the mouse embryo along the anteroposterior (A-P) axis is the migration of the distal visceral endoderm (DVE) toward the future anterior side. Here we report an asymmetry in the mouse embryo at an unexpectedly early stage. The gene for Lefty1, a Nodal antagonist that influences the direction of DVE migration, was found to be asymmetrically expressed in the primitive endoderm of the implanting blastocyst. Lefty1 expression begins randomly in the inner cell mass (ICM) of the blastocyst but is regionalized to one side of the tilted ICM shortly after implantation. Asymmetric expression of Lefty1 can be established by in vitro culture, indicating that it does not require interaction with the uterus. The asymmetric Lefty1 expression is induced by Nodal signaling, although Nodal and genes for its effectors are expressed symmetrically. This asymmetry in molecular patterning of the mouse embryo pushes back the origin of the A-P body axis to the peri-implantation stage.


Subject(s)
Body Patterning/physiology , Cell Polarity/physiology , Embryo Implantation/physiology , Endoderm/physiology , Gene Expression Regulation, Developmental , Membrane Proteins/physiology , Transforming Growth Factor beta/physiology , Animals , Base Sequence , Blastomeres/physiology , Body Patterning/genetics , Cell Polarity/genetics , Cells, Cultured , Embryo Implantation/genetics , In Vitro Techniques , Left-Right Determination Factors , Membrane Proteins/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Nodal Protein , Signal Transduction/physiology , Transforming Growth Factor beta/genetics
17.
Sci Adv ; 6(30): eaba1195, 2020 07.
Article in English | MEDLINE | ID: mdl-32743070

ABSTRACT

Immotile cilia sense extracellular signals such as fluid flow, but whether Ca2+ plays a role in flow sensing has been unclear. Here, we examined the role of ciliary Ca2+ in the flow sensing that initiates the breaking of left-right (L-R) symmetry in the mouse embryo. Intraciliary and cytoplasmic Ca2+ transients were detected in the crown cells at the node. These Ca2+ transients showed L-R asymmetry, which was lost in the absence of fluid flow or the PKD2 channel. Further characterization allowed classification of the Ca2+ transients into two types: cilium-derived, L-R-asymmetric transients (type 1) and cilium-independent transients without an L-R bias (type 2). Type 1 intraciliary transients occurred preferentially at the left posterior region of the node, where L-R symmetry breaking takes place. Suppression of intraciliary Ca2+ transients delayed L-R symmetry breaking. Our results implicate cilium-derived Ca2+ transients in crown cells in initiation of L-R symmetry breaking in the mouse embryo.

18.
Neuron ; 107(1): 82-94.e6, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32330411

ABSTRACT

Patients lacking PYCR2, a mitochondrial enzyme that synthesizes proline, display postnatal degenerative microcephaly with hypomyelination. Here we report the crystal structure of the PYCR2 apo-enzyme and show that a novel germline p.Gly249Val mutation lies at the dimer interface and lowers its enzymatic activity. We find that knocking out Pycr2 in mice phenocopies the human disorder and depletes PYCR1 levels in neural lineages. In situ quantification of neurotransmitters in the brains of PYCR2 mutant mice and patients revealed a signature of encephalopathy driven by excessive cerebral glycine. Mechanistically, we demonstrate that loss of PYCR2 upregulates SHMT2, which is responsible for glycine synthesis. This hyperglycemia could be partially reversed by SHMT2 knockdown, which rescued the axonal beading and neurite lengths of cultured Pycr2 knockout neurons. Our findings identify the glycine metabolic pathway as a possible intervention point to alleviate the neurological symptoms of PYCR2-mutant patients.


Subject(s)
Cerebral Cortex/metabolism , Glycine Hydroxymethyltransferase/metabolism , Glycine/metabolism , Hereditary Central Nervous System Demyelinating Diseases/pathology , Pyrroline Carboxylate Reductases/genetics , Adolescent , Animals , Cerebral Cortex/pathology , Child, Preschool , Female , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Humans , Infant , Male , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Pedigree , Pyrroline Carboxylate Reductases/deficiency
19.
Nat Commun ; 11(1): 5520, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33139725

ABSTRACT

Axonemal dynein ATPases direct ciliary and flagellar beating via adenosine triphosphate (ATP) hydrolysis. The modulatory effect of adenosine monophosphate (AMP) and adenosine diphosphate (ADP) on flagellar beating is not fully understood. Here, we describe a deficiency of cilia and flagella associated protein 45 (CFAP45) in humans and mice that presents a motile ciliopathy featuring situs inversus totalis and asthenospermia. CFAP45-deficient cilia and flagella show normal morphology and axonemal ultrastructure. Proteomic profiling links CFAP45 to an axonemal module including dynein ATPases and adenylate kinase as well as CFAP52, whose mutations cause a similar ciliopathy. CFAP45 binds AMP in vitro, consistent with structural modelling that identifies an AMP-binding interface between CFAP45 and AK8. Microtubule sliding of dyskinetic sperm from Cfap45-/- mice is rescued with the addition of either AMP or ADP with ATP, compared to ATP alone. We propose that CFAP45 supports mammalian ciliary and flagellar beating via an adenine nucleotide homeostasis module.


Subject(s)
Adenine Nucleotides/metabolism , Asthenozoospermia/genetics , Cytoskeletal Proteins/deficiency , Situs Inversus/genetics , Adolescent , Adult , Animals , Asthenozoospermia/pathology , Axoneme/ultrastructure , CRISPR-Cas Systems/genetics , Cilia/metabolism , Cilia/ultrastructure , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Disease Models, Animal , Epididymis/pathology , Female , Flagella/metabolism , Flagella/ultrastructure , Humans , Loss of Function Mutation , Male , Mice , Mice, Knockout , Middle Aged , Planarians/cytology , Planarians/genetics , Planarians/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/pathology , Situs Inversus/diagnostic imaging , Situs Inversus/pathology , Sperm Motility/genetics , Tomography, X-Ray Computed , Exome Sequencing
20.
Elife ; 72018 08 02.
Article in English | MEDLINE | ID: mdl-30070635

ABSTRACT

We have examined the role of Fam60a, a gene highly expressed in embryonic stem cells, in mouse development. Fam60a interacts with components of the Sin3a-Hdac transcriptional corepressor complex, and most Fam60a-/- embryos manifest hypoplasia of visceral organs and die in utero. Fam60a is recruited to the promoter regions of a subset of genes, with the expression of these genes being either up- or down-regulated in Fam60a-/- embryos. The DNA methylation level of the Fam60a target gene Adhfe1 is maintained at embryonic day (E) 7.5 but markedly reduced at E9.5 in Fam60a-/- embryos, suggesting that DNA demethylation is enhanced in the mutant. Examination of genome-wide DNA methylation identified several differentially methylated regions, which were preferentially hypomethylated, in Fam60a-/- embryos. Our data suggest that Fam60a is required for proper embryogenesis, at least in part as a result of its regulation of DNA methylation at specific gene promoters.


Subject(s)
DNA Methylation/genetics , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Animals , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Developmental , Genome , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sin3 Histone Deacetylase and Corepressor Complex
SELECTION OF CITATIONS
SEARCH DETAIL