Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 10688, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37393374

ABSTRACT

The establishment of left-right patterning in mice occurs at a transient structure called the embryonic node or left-right organizer (LRO). Previous analysis of the LRO has proven challenging due to the small cell number and transient nature of this structure. Here, we seek to overcome these difficulties to define the transcriptome of the LRO. Specifically, we used single cell RNA sequencing of 0-1 somite embryos to identify LRO enriched genes which were compared to bulk RNA sequencing of LRO cells isolated by fluorescent activated cell sorting. Gene ontology analysis indicated an enrichment of genes associated with cilia and laterality terms. Furthermore, comparison to previously identified LRO genes identified 127 novel LRO genes, including Ttll3, Syne1 and Sparcl1, for which the expression patterns were validated using whole mount in situ hybridization. This list of novel LRO genes will be a useful resource for further studies on LRO morphogenesis, the establishment of laterality and the genetic causes of heterotaxy.


Subject(s)
Cilia , Transcriptome , Animals , Mice , Cell Count , Cell Separation , Cilia/genetics , RNA , Calcium-Binding Proteins , Extracellular Matrix Proteins
2.
Hum Mol Genet ; 32(14): 2335-2346, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37158461

ABSTRACT

FOXJ1 is expressed in ciliated cells of the airways, testis, oviduct, central nervous system and the embryonic left-right organizer. Ablation or targeted mutation of Foxj1 in mice, zebrafish and frogs results in loss of ciliary motility and/or reduced length and number of motile cilia, affecting the establishment of the left-right axis. In humans, heterozygous pathogenic variants in FOXJ1 cause ciliopathy leading to situs inversus, obstructive hydrocephalus and chronic airway disease. Here, we report a novel truncating FOXJ1 variant (c.784_799dup; p.Glu267Glyfs*12) identified by clinical exome sequencing from a patient with isolated congenital heart defects (CHD) which included atrial and ventricular septal defects, double outlet right ventricle (DORV) and transposition of the great arteries. Functional experiments show that FOXJ1 c.784_799dup; p.Glu267Glyfs*12, unlike FOXJ1, fails to induce ectopic cilia in frog epidermis in vivo or to activate the ADGB promoter, a downstream target of FOXJ1 in cilia, in transactivation assays in vitro. Variant analysis of patients with heterotaxy or heterotaxy-related CHD indicates that pathogenic variants in FOXJ1 are an infrequent cause of heterotaxy. Finally, we characterize embryonic-stage CHD in Foxj1 loss-of-function mice, demonstrating randomized heart looping. Abnormal heart looping includes reversed looping (dextrocardia), ventral looping and no looping/single ventricle hearts. Complex CHDs revealed by histological analysis include atrioventricular septal defects, DORV, single ventricle defects as well as abnormal position of the great arteries. These results indicate that pathogenic variants in FOXJ1 can cause isolated CHD.


Subject(s)
Heart Defects, Congenital , Heart Septal Defects , Heterotaxy Syndrome , Transposition of Great Vessels , Humans , Male , Forkhead Transcription Factors/genetics , Heart Atria , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Transposition of Great Vessels/genetics
3.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638777

ABSTRACT

The mechanisms of neural crest cell induction and specification are highly conserved among vertebrate model organisms, but how similar these mechanisms are in mammalian neural crest cell formation remains open to question. The zinc finger of the cerebellum 1 (ZIC1) transcription factor is considered a core component of the vertebrate gene regulatory network that specifies neural crest fate at the neural plate border. In mouse embryos, however, Zic1 mutation does not cause neural crest defects. Instead, we and others have shown that murine Zic2 and Zic5 mutate to give a neural crest phenotype. Here, we extend this knowledge by demonstrating that murine Zic3 is also required for, and co-operates with, Zic2 and Zic5 during mammalian neural crest specification. At the murine neural plate border (a region of high canonical WNT activity) ZIC2, ZIC3, and ZIC5 function as transcription factors to jointly activate the Foxd3 specifier gene. This function is promoted by SUMOylation of the ZIC proteins at a conserved lysine immediately N-terminal of the ZIC zinc finger domain. In contrast, in the lateral regions of the neurectoderm (a region of low canonical WNT activity) basal ZIC proteins act as co-repressors of WNT/TCF-mediated transcription. Our work provides a mechanism by which mammalian neural crest specification is restricted to the neural plate border. Furthermore, given that WNT signaling and SUMOylation are also features of non-mammalian neural crest specification, it suggests that mammalian neural crest induction shares broad conservation, but altered molecular detail, with chicken, zebrafish, and Xenopus neural crest induction.


Subject(s)
Embryo, Mammalian/embryology , Neural Crest/metabolism , Sumoylation , Transcription Factors/metabolism , Transcription, Genetic , Wnt Signaling Pathway , Animals , Embryo, Mammalian/cytology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mice, Transgenic , Neural Crest/cytology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
4.
Hum Mol Genet ; 30(24): 2402-2415, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34274973

ABSTRACT

Loss of function of ZIC3 causes heterotaxy (OMIM #306955), a disorder characterized by organ laterality defects including complex heart defects. Studies using Zic3 mutant mice have demonstrated that loss of Zic3 causes heterotaxy due to defects in establishment of left-right (LR) signaling, but the mechanistic basis for these defects remains unknown. Here, we demonstrate Zic3 null mice undergo cilia positioning defects at the embryonic node consistent with impaired planar cell polarity (PCP). Cell-based assays demonstrate that ZIC3 must enter the nucleus to regulate PCP and identify multiple critical ZIC3 domains required for regulation of PCP signaling. Furthermore, we show that Zic3 displays a genetic interaction with the PCP membrane protein Vangl2 and the PCP effector genes Rac1 and Daam1 resulting in increased frequency and severity of neural tube and heart defects. Gene and protein expression analyses indicate that Zic3 null embryos display disrupted expression of PCP components and reduced phosphorylation of the core PCP protein DVL2 at the time of LR axis determination. These results demonstrate that ZIC3 interacts with PCP signaling during early development, identifying a novel role for this transcription factor, and adding additional evidence about the importance of PCP function for normal LR patterning and subsequent heart development.


Subject(s)
Heterotaxy Syndrome , Homeodomain Proteins , Neural Tube Defects , Transcription Factors , Animals , Cell Polarity/genetics , Heterotaxy Syndrome/genetics , Homeodomain Proteins/genetics , Mice , Microfilament Proteins/metabolism , Neural Tube , Neural Tube Defects/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism
5.
J Cell Sci ; 134(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33771929

ABSTRACT

Zinc finger of the cerebellum (Zic) proteins act as classic transcription factors to promote transcription of the Foxd3 gene during neural crest cell specification. Additionally, they can act as co-factors that bind proteins from the T-cell factor/lymphoid enhancing factor (TCF/LEF) family (TCFs) to repress WNT-ß-catenin-dependent transcription without contacting DNA. Here, we show that ZIC activity at the neural plate border is influenced by WNT-dependent SUMOylation. In the presence of high canonical WNT activity, a lysine residue within the highly conserved zinc finger N-terminally conserved (ZF-NC) domain of ZIC5 is SUMOylated, which reduces formation of the ZIC-TCF co-repressor complex and shifts the balance towards transcription factor function. The modification is crucial in vivo, as a ZIC5 SUMO-incompetent mouse strain exhibits neural crest specification defects. This work reveals the function of the ZF-NC domain within ZIC, provides in vivo validation of target protein SUMOylation and demonstrates that WNT-ß-catenin signalling directs transcription at non-TCF DNA-binding sites. Furthermore, it can explain how WNT signals convert a broad region of Zic ectodermal expression into a restricted region of neural crest cell specification.


Subject(s)
Neural Crest , Sumoylation , Animals , Cell Differentiation , Mice , Neural Crest/metabolism , TCF Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
6.
Sci Rep ; 10(1): 13130, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753700

ABSTRACT

The ZIC proteins are a family of transcription regulators with a well-defined zinc finger DNA-binding domain and there is evidence that they elicit functional DNA binding at a ZIC DNA binding site. Little is known, however, regarding domains within ZIC proteins that confer trans-activation or -repression. To address this question, a new cell-based trans-activation assay system suitable for ZIC proteins in HEK293T cells was constructed. This identified two previously unannotated evolutionarily conserved regions of ZIC3 that are necessary for trans-activation. These domains are found in all Subclass A ZIC proteins, but not in the Subclass B proteins. Additionally, the Subclass B proteins fail to elicit functional binding at a multimerised ZIC DNA binding site. All ZIC proteins, however, exhibit functional binding when the ZIC DNA binding site is embedded in a multiple transcription factor locus derived from ZIC target genes in the mouse genome. This ability is due to several domains, some of which are found in all ZIC proteins, that exhibit context dependent trans-activation or -repression activity. This knowledge is valuable for assessing the likely pathogenicity of variant ZIC proteins associated with human disorders and for determining factors that influence functional transcription factor binding.


Subject(s)
Gene Expression Regulation , Genes, Reporter , Response Elements , Transcription Factors , Transcription, Genetic , HEK293 Cells , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Adv Exp Med Biol ; 1046: 301-327, 2018.
Article in English | MEDLINE | ID: mdl-29442328

ABSTRACT

Mutation of ZIC3 causes X-linked heterotaxy, a syndrome in which the laterality of internal organs is disrupted. Analysis of model organisms and gene expression during early development suggests ZIC3-related heterotaxy occurs due to defects at the earliest stage of left-right axis formation. Although there are data to support abnormalities of the node and cilia as underlying causes, it is unclear at the molecular level why loss of ZIC3 function causes such these defects. ZIC3 has putative roles in a number of developmental signalling pathways that have distinct roles in establishing the left-right axis. This complicates the understanding of the mechanistic basis of Zic3 in early development and left-right patterning. Here we summarise our current understanding of ZIC3 function and describe the potential role ZIC3 plays in important signalling pathways and their links to heterotaxy.


Subject(s)
Dextrocardia , Genetic Diseases, X-Linked , Heterotaxy Syndrome , Homeodomain Proteins , Mutation , Signal Transduction/genetics , Transcription Factors , Animals , Dextrocardia/embryology , Dextrocardia/genetics , Genetic Diseases, X-Linked/embryology , Genetic Diseases, X-Linked/genetics , Heterotaxy Syndrome/embryology , Heterotaxy Syndrome/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Dis Model Mech ; 6(3): 755-67, 2013 May.
Article in English | MEDLINE | ID: mdl-23471918

ABSTRACT

The ZIC transcription factors are key mediators of embryonic development and ZIC3 is the gene most commonly associated with situs defects (heterotaxy) in humans. Half of patient ZIC3 mutations introduce a premature termination codon (PTC). In vivo, PTC-containing transcripts might be targeted for nonsense-mediated decay (NMD). NMD efficiency is known to vary greatly between transcripts, tissues and individuals and it is possible that differences in survival of PTC-containing transcripts partially explain the striking phenotypic variability that characterizes ZIC3-associated congenital defects. For example, the PTC-containing transcripts might encode a C-terminally truncated protein that retains partial function or that dominantly interferes with other ZIC family members. Here we describe the katun (Ka) mouse mutant, which harbours a mutation in the Zic3 gene that results in a PTC. At the time of axis formation there is no discernible decrease in this PTC-containing transcript in vivo, indicating that the mammalian Zic3 transcript is relatively insensitive to NMD, prompting the need to re-examine the molecular function of the truncated proteins predicted from human studies and to determine whether the N-terminal portion of ZIC3 possesses dominant-negative capabilities. A combination of in vitro studies and analysis of the Ka phenotype indicate that it is a null allele of Zic3 and that the N-terminal portion of ZIC3 does not encode a dominant-negative molecule. Heterotaxy in patients with PTC-containing ZIC3 transcripts probably arises due to loss of ZIC3 function alone.


Subject(s)
Codon, Nonsense/genetics , Heterotaxy Syndrome/embryology , Heterotaxy Syndrome/genetics , Homeodomain Proteins/metabolism , Nonsense Mediated mRNA Decay/genetics , Transcription Factors/metabolism , Alleles , Animals , Base Sequence , Cell Nucleus/metabolism , Diffusion , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endoderm/abnormalities , Endoderm/embryology , Gastrulation/genetics , Homeodomain Proteins/genetics , Humans , Mesoderm/abnormalities , Mesoderm/embryology , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Organogenesis/genetics , Protein Stability , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription, Genetic , beta Catenin/metabolism
9.
Int J Biochem Cell Biol ; 44(11): 2065-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22964024

ABSTRACT

The Zic genes encode zinc finger containing proteins that can bind proteins and DNA. The understanding of Zic molecular networks has been hampered by functional redundancy amongst family members, and because their loss-of-function phenotypes are indicative of a role in many signalling pathways. Recently molecular evidence has emerged confirming the pleiotropic nature of these proteins: they act both as classical transcription factors and as co-factors to directly and indirectly influence gene expression. It has long been known that germ-line mutation of the Zic genes in human and mouse causes a range of congenital disorders. Recently connections between Zic proteins and stem cell function have also emerged suggesting a role in adult onset diseases. The immediate challenge is to determine when and where these proteins act as transcription factors/co-factors during development and disease and how the switch between these roles is controlled.


Subject(s)
Cerebellum/metabolism , Transcription Factors/metabolism , Zinc Fingers , Animals , Disease , Gene Expression Regulation , Humans , Models, Biological , Transcription Factors/chemistry , Transcription Factors/genetics , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...