Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 665
Filter
Add more filters

Publication year range
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.
Cell ; 148(1-2): 273-84, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265416

ABSTRACT

Secreted signals, known as morphogens, provide the positional information that organizes gene expression and cellular differentiation in many developing tissues. In the vertebrate neural tube, Sonic Hedgehog (Shh) acts as a morphogen to control the pattern of neuronal subtype specification. Using an in vivo reporter of Shh signaling, mouse genetics, and systems modeling, we show that a spatially and temporally changing gradient of Shh signaling is interpreted by the regulatory logic of a downstream transcriptional network. The design of the network, which links three transcription factors to Shh signaling, is responsible for differential spatial and temporal gene expression. In addition, the network renders cells insensitive to fluctuations in signaling and confers hysteresis--memory of the signal. Our findings reveal that morphogen interpretation is an emergent property of the architecture of a transcriptional network that provides robustness and reliability to tissue patterning.


Subject(s)
Gene Regulatory Networks , Hedgehog Proteins/metabolism , Neural Tube/metabolism , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Eye Proteins/genetics , Hedgehog Proteins/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Oligodendrocyte Transcription Factor 2 , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Transcription Factors/metabolism , Zebrafish Proteins , Zinc Finger Protein Gli3
3.
Proc Natl Acad Sci U S A ; 121(28): e2320070121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968120

ABSTRACT

Hedgehog (Hh) signaling, an evolutionarily conserved pathway, plays an essential role in development and tumorigenesis, making it a promising drug target. Multiple negative regulators are known to govern Hh signaling; however, how activated Smoothened (SMO) participates in the activation of downstream GLI2 and GLI3 remains unclear. Herein, we identified the ciliary kinase DYRK2 as a positive regulator of the GLI2 and GLI3 transcription factors for Hh signaling. Transcriptome and interactome analyses demonstrated that DYRK2 phosphorylates GLI2 and GLI3 on evolutionarily conserved serine residues at the ciliary base, in response to activation of the Hh pathway. This phosphorylation induces the dissociation of GLI2/GLI3 from suppressor, SUFU, and their translocation into the nucleus. Loss of Dyrk2 in mice causes skeletal malformation, but neural tube development remains normal. Notably, DYRK2-mediated phosphorylation orchestrates limb development by controlling cell proliferation. Taken together, the ciliary kinase DYRK2 governs the activation of Hh signaling through the regulation of two processes: phosphorylation of GLI2 and GLI3 downstream of SMO and cilia formation. Thus, our findings of a unique regulatory mechanism of Hh signaling expand understanding of the control of Hh-associated diseases.


Subject(s)
Dyrk Kinases , Hedgehog Proteins , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Signal Transduction , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3 , Animals , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Humans , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Cell Proliferation , Cilia/metabolism , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Nuclear Proteins , Repressor Proteins
4.
Proc Natl Acad Sci U S A ; 121(20): e2321711121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713624

ABSTRACT

During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cerebral Cortex , ErbB Receptors , Hedgehog Proteins , Nerve Tissue Proteins , Neural Stem Cells , Neurogenesis , Oligodendrocyte Transcription Factor 2 , PAX6 Transcription Factor , Animals , Neurogenesis/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Mice , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendrocyte Transcription Factor 2/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Neuroglia/metabolism , Neuroglia/cytology , Gene Expression Regulation, Developmental , Signal Transduction , Olfactory Bulb/metabolism , Olfactory Bulb/cytology , Cell Lineage , Humans
5.
J Med Genet ; 61(7): 633-644, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38531627

ABSTRACT

BACKGROUND: Ellis-van Creveld syndrome (EvC) is a recessive disorder characterised by acromesomelic limb shortening, postaxial polydactyly, nail-teeth dysplasia and congenital cardiac defects, primarily caused by pathogenic variants in EVC or EVC2. Weyers acrofacial dysostosis (WAD) is an ultra-rare dominant condition allelic to EvC. The present work aimed to enhance current knowledge on the clinical manifestations of EvC and WAD and broaden their mutational spectrum. METHODS: We conducted molecular studies in 46 individuals from 43 unrelated families with a preliminary clinical diagnosis of EvC and 3 affected individuals from a family with WAD and retrospectively analysed clinical data. The deleterious effect of selected variants of uncertain significance was evaluated by cellular assays. MAIN RESULTS: We identified pathogenic variants in EVC/EVC2 in affected individuals from 41 of the 43 families with EvC. Patients from each of the two remaining families were found with a homozygous splicing variant in WDR35 and a de novo heterozygous frameshift variant in GLI3, respectively. The phenotype of these patients showed a remarkable overlap with EvC. A novel EVC2 C-terminal truncating variant was identified in the family with WAD. Deep phenotyping of the cohort recapitulated 'classical EvC findings' in the literature and highlighted findings previously undescribed or rarely described as part of EvC. CONCLUSIONS: This study presents the largest cohort of living patients with EvC to date, contributing to better understanding of the full clinical spectrum of EvC. We also provide comprehensive information on the EVC/EVC2 mutational landscape and add GLI3 to the list of genes associated with EvC-like phenotypes.


Subject(s)
Ellis-Van Creveld Syndrome , Pedigree , Phenotype , Humans , Ellis-Van Creveld Syndrome/genetics , Ellis-Van Creveld Syndrome/pathology , Male , Female , Child , Membrane Proteins/genetics , Mutation , Child, Preschool , Zinc Finger Protein Gli3/genetics , Adolescent , Adult , Nerve Tissue Proteins/genetics , Cohort Studies , Infant , Proteins/genetics , Retrospective Studies , Intercellular Signaling Peptides and Proteins
6.
PLoS Genet ; 18(7): e1010315, 2022 07.
Article in English | MEDLINE | ID: mdl-35867772

ABSTRACT

Proper Hedgehog (HH) signaling is essential for embryonic development, while aberrant HH signaling drives pediatric and adult cancers. HH signaling is frequently dysregulated in pancreatic cancer, yet its role remains controversial, with both tumor-promoting and tumor-restraining functions reported. Notably, the GLI family of HH transcription factors (GLI1, GLI2, GLI3), remain largely unexplored in pancreatic cancer. We therefore investigated the individual and combined contributions of GLI1-3 to pancreatic cancer progression. At pre-cancerous stages, fibroblast-specific Gli2/Gli3 deletion decreases immunosuppressive macrophage infiltration and promotes T cell infiltration. Strikingly, combined loss of Gli1/Gli2/Gli3 promotes macrophage infiltration, indicating that subtle changes in Gli expression differentially regulate immune infiltration. In invasive tumors, Gli2/Gli3 KO fibroblasts exclude immunosuppressive myeloid cells and suppress tumor growth by recruiting natural killer cells. Finally, we demonstrate that fibroblasts directly regulate macrophage and T cell migration through the expression of Gli-dependent cytokines. Thus, the coordinated activity of GLI1-3 directs the fibroinflammatory response throughout pancreatic cancer progression.


Subject(s)
Hedgehog Proteins , Pancreatic Neoplasms , Adult , Child , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/genetics , Pregnancy , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli3/genetics
7.
Dev Biol ; 504: 128-136, 2023 12.
Article in English | MEDLINE | ID: mdl-37805104

ABSTRACT

Transcriptional responses to the Hedgehog (HH) signaling pathway are primarily modulated by GLI repression in the mouse limb. Previous studies suggested a role for the BAF chromatin remodeling complex in mediating GLI repression. Consistent with this possibility, the core BAF complex protein SMARCC1 is present at most active limb enhancers including the majority of GLI enhancers. However, in contrast to GLI repression which reduces chromatin accessibility, SMARCC1 maintains chromatin accessibility at most enhancers, including those bound by GLI. Moreover, SMARCC1 binding at GLI-regulated enhancers occurs independently of GLI3. Consistent with previous studies, some individual GLI target genes are mis-regulated in Smarcc1 conditional knockouts, though most GLI target genes are unaffected. Moreover, SMARCC1 is not necessary for mediating constitutive GLI repression in HH mutant limb buds. We conclude that SMARCC1 does not mediate GLI3 repression, which we propose utilizes alternative chromatin remodeling complexes.


Subject(s)
Chromatin , Limb Buds , Animals , Mice , Chromatin/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Limb Buds/metabolism , Nerve Tissue Proteins/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli3/metabolism
8.
J Med Genet ; 60(5): 505-510, 2023 05.
Article in English | MEDLINE | ID: mdl-36411030

ABSTRACT

Many genetic testing methodologies are biased towards picking up structural variants (SVs) that alter copy number. Copy-neutral rearrangements such as inversions are therefore likely to suffer from underascertainment. In this study, manual review prompted by a virtual multidisciplinary team meeting and subsequent bioinformatic prioritisation of data from the 100K Genomes Project was performed across 43 genes linked to well-characterised skeletal disorders. Ten individuals from three independent families were found to harbour diagnostic inversions. In two families, inverted segments of 1.2/14.8 Mb unequivocally disrupted GLI3 and segregated with skeletal features consistent with Greig cephalopolysyndactyly syndrome. For one family, phenotypic blending was due to the opposing breakpoint lying ~45 kb from HOXA13 In the third family, long suspected to have Marfan syndrome, a 2.0 Mb inversion disrupting FBN1 was identified. These findings resolved lengthy diagnostic odysseys of 9-20 years and highlight the importance of direct interaction between clinicians and data-analysts. These exemplars of a rare mutational class inform future SV prioritisation strategies within the NHS Genomic Medicine Service and similar genome sequencing initiatives. In over 30 years since these two disease-gene associations were identified, large inversions have yet to be described and so our results extend the mutational spectra linked to these conditions.


Subject(s)
Bone Diseases, Developmental , Chromosome Inversion , Humans , Base Sequence , Bone Diseases, Developmental/diagnosis , Bone Diseases, Developmental/genetics , Chromosome Inversion/genetics , Chromosome Mapping , Fibrillin-1/genetics , Genetic Testing , Mutation , Nerve Tissue Proteins/genetics , Zinc Finger Protein Gli3/genetics
9.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Article in English | MEDLINE | ID: mdl-34750251

ABSTRACT

One of the central problems of vertebrate evolution is understanding the relationship among the distal portions of fins and limbs. Lacking comparable morphological markers of these regions in fish and tetrapods, these relationships have remained uncertain for the past century and a half. Here we show that Gli3 functions in controlling the proliferative expansion of distal progenitors are shared among dorsal and paired fins as well as tetrapod limbs. Mutant knockout gli3 fins in medaka (Oryzias latipes) form multiple radials and rays, in a pattern reminiscent of the polydactyly observed in Gli3-null mutant mice. In limbs, Gli3 controls both anterior-posterior patterning and cell proliferation, two processes that can be genetically uncoupled. In situ hybridization, quantification of proliferation markers, and analysis of regulatory regions reveal that in paired and dorsal fins, gli3 plays a main role in controlling proliferation but not in patterning. Moreover, gli3 down-regulation in shh mutant fins rescues fin loss in a manner similar to how Gli3 deficiency restores digits in the limbs of Shh mutant mouse embryos. We hypothesize that the Gli3/Shh gene pathway preceded the origin of paired appendages and was originally involved in modulating cell proliferation. Accordingly, the distal regions of dorsal fins, paired fins, and limbs retain a deep regulatory and functional homology that predates the origin of paired appendages.


Subject(s)
Animal Fins/growth & development , Gene Regulatory Networks/genetics , Nerve Tissue Proteins/genetics , Oryzias/genetics , Zinc Finger Protein Gli3/genetics , Animals , Biological Evolution , Body Patterning/genetics , Cell Proliferation/genetics , Extremities/growth & development , Fish Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Mice
10.
PLoS Genet ; 17(12): e1009982, 2021 12.
Article in English | MEDLINE | ID: mdl-34928956

ABSTRACT

Sonic Hedgehog/GLI3 signaling is critical in regulating digit number, such that Gli3-deficiency results in polydactyly and Shh-deficiency leads to digit number reductions. SHH/GLI3 signaling regulates cell cycle factors controlling mesenchymal cell proliferation, while simultaneously regulating Grem1 to coordinate BMP-induced chondrogenesis. SHH/GLI3 signaling also coordinates the expression of additional genes, however their importance in digit formation remain unknown. Utilizing genetic and molecular approaches, we identified HES1 as a downstream modifier of the SHH/GLI signaling axis capable of inducing preaxial polydactyly (PPD), required for Gli3-deficient PPD, and capable of overcoming digit number constraints of Shh-deficiency. Our data indicate that HES1, a direct SHH/GLI signaling target, induces mesenchymal cell proliferation via suppression of Cdkn1b, while inhibiting chondrogenic genes and the anterior autopod boundary regulator, Pax9. These findings establish HES1 as a critical downstream effector of SHH/GLI3 signaling in the development of PPD.


Subject(s)
Hedgehog Proteins/genetics , Nerve Tissue Proteins/genetics , PAX9 Transcription Factor/genetics , Polydactyly/genetics , Thumb/abnormalities , Transcription Factor HES-1/genetics , Zinc Finger Protein Gli3/genetics , Animals , Cell Division/genetics , Cell Proliferation/genetics , Chondrogenesis/genetics , Chromatin/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Disease Models, Animal , Humans , Limb Buds/growth & development , Limb Buds/metabolism , Mesoderm/growth & development , Mice , Polydactyly/pathology , Thumb/pathology
11.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731849

ABSTRACT

Tumors of the head and neck, more specifically the squamous cell carcinoma, often show upregulation of the Hedgehog signaling pathway. However, almost nothing is known about its role in the sinonasal adenocarcinoma, either in intestinal or non-intestinal subtypes. In this work, we have analyzed immunohistochemical staining of six Hedgehog pathway proteins, sonic Hedgehog (SHH), Indian Hedgehog (IHH), Patched1 (PTCH1), Gli family zinc finger 1 (GLI1), Gli family zinc finger 2 (GLI2), and Gli family zinc finger 3 (GLI3), on 21 samples of sinonasal adenocarcinoma and compared them with six colon adenocarcinoma and three salivary gland tumors, as well as with matching healthy tissue, where available. We have detected GLI2 and PTCH1 in the majority of samples and also GLI1 in a subset of samples, while GLI3 and the ligands SHH and IHH were generally not detected. PTCH1 pattern of staining shows an interesting pattern, where healthy samples are mostly positive in the stromal compartment, while the signal shifts to the tumor compartment in tumors. This, taken together with a stronger signal of GLI2 in tumors compared to non-tumor tissues, suggests that the Hedgehog pathway is indeed activated in sinonasal adenocarcinoma. As Hedgehog pathway inhibitors are being tested in combination with other therapies for head and neck squamous cell carcinoma, this could provide a therapeutic option for patients with sinonasal adenocarcinoma as well.


Subject(s)
Adenocarcinoma , Hedgehog Proteins , Immunohistochemistry , Signal Transduction , Zinc Finger Protein Gli2 , Humans , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Male , Female , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Middle Aged , Pilot Projects , Aged , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Paranasal Sinus Neoplasms/metabolism , Paranasal Sinus Neoplasms/pathology , Adult , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins , Nuclear Proteins
12.
Am J Hum Genet ; 106(6): 779-792, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32413283

ABSTRACT

The evolutionarily conserved hedgehog (Hh) pathway is essential for organogenesis and plays critical roles in postnatal tissue maintenance and renewal. A unique feature of the vertebrate Hh pathway is that signal transduction requires the primary cilium (PC) where major pathway components are dynamically enriched. These factors include smoothened (SMO) and patched, which constitute the core reception system for sonic hedgehog (SHH) as well as GLI transcription factors, the key mediators of the pathway. Here, we report bi-allelic loss-of-function variations in SMO in seven individuals from five independent families; these variations cause a wide phenotypic spectrum of developmental anomalies affecting the brain (hypothalamic hamartoma and microcephaly), heart (atrioventricular septal defect), skeleton (postaxial polydactyly, narrow chest, and shortening of long bones), and enteric nervous system (aganglionosis). Cells derived from affected individuals showed normal ciliogenesis but severely altered Hh-signal transduction as a result of either altered PC trafficking or abnormal activation of the pathway downstream of SMO. In addition, Hh-independent GLI2 accumulation at the PC tip in cells from the affected individuals suggests a potential function of SMO in regulating basal ciliary trafficking of GLI2 when the pathway is off. Thus, loss of SMO function results in abnormal PC dynamics of key components of the Hh signaling pathway and leads to a large continuum of malformations in humans.


Subject(s)
Alleles , Developmental Disabilities/genetics , Hedgehog Proteins/metabolism , Signal Transduction , Smoothened Receptor/genetics , Base Sequence , Child , Child, Preschool , Cilia/physiology , Female , Humans , Infant , Male , Models, Molecular , Neoplasms/genetics , Nerve Tissue Proteins , Nuclear Proteins , Pedigree , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
13.
Development ; 147(3)2020 02 03.
Article in English | MEDLINE | ID: mdl-31932349

ABSTRACT

Cerebellar granule cell (GC) development relies on precise regulation of sonic hedgehog (Shh)-Gli signalling activity, failure of which is associated with motor disorders and medulloblastoma. Mutations in the pathway regulator suppressor of fused (Sufu), which modulates Gli activators and repressors, are linked to cerebellar dysfunction and tumourigenesis. The mechanism by which Sufu calibrates Shh signalling in GCs is unknown. Math1-Cre-mediated deletion of Sufu in mouse GC progenitors (GCPs) demonstrated that Sufu restricts GCP proliferation and promotes cell cycle exit, by promoting expression of Gli3R and suppressing Gli2 levels. Sufu is also required to promote a high threshold of pathway activity in GCPs. Remarkably, central cerebellar lobules are more deleteriously impacted by Sufu deletion, but are less sensitive to downstream genetic manipulations to reduce Gli2 expression or overexpress a Gli3R mimic, compared with anterior lobules. Transcriptome sequencing uncovered new Sufu targets, especially Fgf8, which is upregulated in Sufu-mutant GCPs. We demonstrate that Fgf8 is necessary and sufficient to drive Sufu-mutant GCP proliferation. This study reveals new insights into the spatial and temporal regulation of cerebellar Shh-Gli signalling, while uncovering new targets, such as Fgf8.


Subject(s)
Cell Proliferation/genetics , Cerebellum/cytology , Fibroblast Growth Factor 8/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Repressor Proteins/metabolism , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli3/metabolism , Animals , Cell Cycle/genetics , Cerebellum/growth & development , Female , Fibroblast Growth Factor 8/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Transcriptome , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli3/genetics
14.
Development ; 147(3)2020 02 03.
Article in English | MEDLINE | ID: mdl-31964774

ABSTRACT

Sonic hedgehog (Shh) signal transduction specifies ventral cell fates in the neural tube and is mediated by the Gli transcription factors that play both activator (GliA) and repressor (GliR) roles. Cilia are essential for Shh signal transduction and the ciliary phosphatidylinositol phosphatase Inpp5e is linked to Shh regulation. In the course of a forward genetic screen for recessive mouse mutants, we identified a functional null allele of inositol polyphosphate-5-phosphatase E (Inpp5e), ridge top (rdg), with expanded ventral neural cell fates at E10.5. By E12.5, Inpp5erdg/rdg embryos displayed normal neural patterning and this correction over time required Gli3, the predominant repressor in neural patterning. Inpp5erdg function largely depended on the presence of cilia and on smoothened, the obligate transducer of Shh signaling, indicating that Inpp5e functions within the cilium to regulate the pathway. These data indicate that Inpp5e plays a more complicated role in Shh signaling than previously appreciated. We propose that Inpp5e attenuates Shh signaling in the neural tube through regulation of the relative timing of GliA and GliR production, which is important in understanding how the duration of Shh signaling regulates neural tube patterning.


Subject(s)
Cilia/metabolism , Hedgehog Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction/genetics , Alleles , Animals , Body Patterning/genetics , Embryo, Mammalian/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Tube/metabolism , Phosphoric Monoester Hydrolases/genetics , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli3/metabolism
15.
Proc Natl Acad Sci U S A ; 117(2): 1090-1096, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31896583

ABSTRACT

In the tetrapod limb, the digits (fingers or toes) are the elements most subject to morphological diversification in response to functional adaptations. However, despite their functional importance, the mechanisms controlling digit morphology remain poorly understood. Here we have focused on understanding the special morphology of the thumb (digit 1), the acquisition of which was an important adaptation of the human hand. To this end, we have studied the limbs of the Hoxa13 mouse mutant that specifically fail to form digit 1. We show that, consistent with the role of Hoxa13 in Hoxd transcriptional regulation, the expression of Hoxd13 in Hoxa13 mutant limbs does not extend into the presumptive digit 1 territory, which is therefore devoid of distal Hox transcripts, a circumstance that can explain its agenesis. The loss of Hoxd13 expression, exclusively in digit 1 territory, correlates with increased Gli3 repressor activity, a Hoxd negative regulator, resulting from increased Gli3 transcription that, in turn, is due to the release from the negative modulation exerted by Hox13 paralogs on Gli3 regulatory sequences. Our results indicate that Hoxa13 acts hierarchically to initiate the formation of digit 1 by reducing Gli3 transcription and by enabling expansion of the 5'Hoxd second expression phase, thereby establishing anterior-posterior asymmetry in the handplate. Our work uncovers a mutual antagonism between Gli3 and Hox13 paralogs that has important implications for Hox and Gli3 gene regulation in the context of development and evolution.


Subject(s)
Extremities/growth & development , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Zinc Finger Protein Gli3/metabolism , Animals , Body Patterning , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Transcriptome , Zinc Finger Protein Gli3/genetics
16.
PLoS Genet ; 16(6): e1008810, 2020 06.
Article in English | MEDLINE | ID: mdl-32497091

ABSTRACT

Urogenital tract abnormalities are among the most common congenital defects in humans. Male urogenital development requires Hedgehog-GLI signaling and testicular hormones, but how these pathways interact is unclear. We found that Gli3XtJ mutant mice exhibit cryptorchidism and hypospadias due to local effects of GLI3 loss and systemic effects of testicular hormone deficiency. Fetal Leydig cells, the sole source of these hormones in developing testis, were reduced in numbers in Gli3XtJ testes, and their functional identity diminished over time. Androgen supplementation partially rescued testicular descent but not hypospadias in Gli3XtJ mutants, decoupling local effects of GLI3 loss from systemic effects of androgen insufficiency. Reintroduction of GLI3 activator (GLI3A) into Gli3XtJ testes restored expression of Hedgehog pathway and steroidogenic genes. Together, our results show a novel function for the activated form of GLI3 that translates Hedgehog signals to reinforce fetal Leydig cell identity and stimulate timely INSL3 and testosterone synthesis in the developing testis. In turn, exquisite timing and concentrations of testosterone are required to work alongside local GLI3 activity to control development of a functionally integrated male urogenital tract.


Subject(s)
Cryptorchidism/genetics , Gene Expression Regulation, Developmental , Leydig Cells/pathology , Nerve Tissue Proteins/metabolism , Sex Differentiation/genetics , Zinc Finger Protein Gli3/metabolism , Animals , Cryptorchidism/pathology , Disease Models, Animal , Hedgehog Proteins/metabolism , Humans , Insulin/metabolism , Leydig Cells/metabolism , Male , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Proteins/metabolism , Signal Transduction/genetics , Testosterone/metabolism , Zinc Finger Protein Gli3/genetics
17.
Genes Dev ; 29(3): 262-76, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644602

ABSTRACT

Binding of the Hedgehog (Hh) protein signal to its receptor, Patched, induces accumulation of the seven-pass transmembrane protein Smoothened (Smo) within the primary cilium and of the zinc finger transcription factor Gli2 at the ciliary tip, resulting ultimately in Gli-mediated changes in nuclear gene expression. However, the mechanism by which pathway activation is communicated from Smo to Gli2 is not known. In an effort to elucidate this mechanism, we identified Dlg5 (Discs large, homolog 5) in a biochemical screen for proteins that preferentially interact with activated Smo. We found that disruption of Smo-Dlg5 interactions or depletion of endogenous Dlg5 leads to diminished Hh pathway response without a significant impact on Smo ciliary accumulation. We also found that Dlg5 is localized at the basal body, where it associates with another pathway component, Kif7. We show that Dlg5 is required for Hh-induced enrichment of Kif7 and Gli2 at the tip of the cilium but is dispensable for Gpr161 exit from the cilium and the consequent suppression of Gli3 processing into its repressor form. Our findings suggest a bifurcation of Smo activity in Hh response, with a Dlg5-independent arm for suppression of Gli repressor formation and a second arm involving Smo interaction with Dlg5 for Gli activation.


Subject(s)
Guanylate Kinases/metabolism , Hedgehog Proteins/metabolism , Membrane Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Basal Bodies/metabolism , Cell Line , Cilia/metabolism , Guanylate Kinases/genetics , HEK293 Cells , Humans , Kinesins/metabolism , Kruppel-Like Transcription Factors/metabolism , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Transport , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
18.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 40(1): 92-95, 2023 Jan 10.
Article in Zh | MEDLINE | ID: mdl-36585009

ABSTRACT

OBJECTIVE: To explore the clinical and genetic characteristics of a child with Pallister-Hall syndrome (PHS). METHODS: DNA was extracted from peripheral blood sample from the child and subjected to whole exome sequencing. Suspected variants were verified by Sanger sequencing of his family members. RESULTS: Genetic testing revealed that the child has harbored a heterozygous c.3320_3330delGGTACGAGCAG (p.G1107Afs×18) variant of the GLI3 gene. Neither parent was found to carry the same variant. CONCLUSION: The c.3320_3330delGGTACGAGCAG (p.G1107Afs×18) frameshift variant of the GLI3 gene probably underlay the pathogenesis of PHS in this child. Genetic testing should be considered for patients featuring hypothalamic hamartoma and central polydactyly.


Subject(s)
Hamartoma , Pallister-Hall Syndrome , Polydactyly , Humans , Child , Pallister-Hall Syndrome/genetics , Kruppel-Like Transcription Factors/genetics , Zinc Finger Protein Gli3/genetics , Polydactyly/genetics , Hamartoma/genetics , Hamartoma/pathology , Nerve Tissue Proteins/genetics
19.
Am J Med Genet C Semin Med Genet ; 190(3): 264-278, 2022 09.
Article in English | MEDLINE | ID: mdl-36165461

ABSTRACT

Pallister-Hall syndrome (PHS) is a rare autosomal dominant disease diagnosed by the presence of hypothalamic hamartoma, mesoaxial polydactyly and a truncating variant in the middle third of the GLI-Kruppel family member 3 (GLI3) gene. PHS may also include a wide range of clinical phenotypes affecting multiple organ systems including congenital anomalies of the kidney and urinary tract (CAKUT). The observed clinical phenotypes are consistent with the essential role of GLI3, a transcriptional effector in the hedgehog (Hh) signaling pathway, in organogenesis. However, the mechanisms by which truncation of GLI3 in PHS results in such a variety of clinical phenotypes with variable severity, even within the same organ, remain unclear. In this study we focus on presentation of CAKUT in PHS. A systematic analysis of reported PHS patients (n = 78) revealed a prevalence of 26.9% (21/78) of CAKUT. Hypoplasia (± dysplasia) and agenesis were the two main types of CAKUT; bilateral and unilateral CAKUT were reported with equal frequency. Examination of clinical phenotypes with CAKUT revealed a significant association between CAKUT and craniofacial defects, bifid epiglottis and a Disorder of Sex Development, specifically affecting external genitalia. Lastly, we determined that PHS patients with CAKUT predominately had substitution type variants (as opposed to deletion type variants in non-CAKUT PHS patients) in the middle third of the GLI3 gene. These results provide a foundation for future work aimed at uncovering the molecular mechanisms by which variant GLI3 result in the wide range and severity of clinical features observed in PHS.


Subject(s)
Abnormalities, Multiple , Pallister-Hall Syndrome , Urinary Tract , Humans , Pallister-Hall Syndrome/diagnosis , Pallister-Hall Syndrome/genetics , Zinc Finger Protein Gli3/genetics , Kruppel-Like Transcription Factors/genetics , Abnormalities, Multiple/genetics , Nerve Tissue Proteins/genetics , Hedgehog Proteins , Kidney
20.
Development ; 146(17)2019 09 12.
Article in English | MEDLINE | ID: mdl-31444216

ABSTRACT

Dorsal-ventral pattern formation of the neural tube is regulated by temporal and spatial activities of extracellular signalling molecules. Sonic hedgehog (Shh) assigns ventral neural subtypes via activation of the Gli transcription factors. Shh activity in the neural progenitor cells changes dynamically during differentiation, but the mechanisms regulating this dynamicity are not fully understood. Here, we show that temporal change of intracellular cAMP levels confers the temporal Shh signal, and the purinergic G-protein-coupled receptor GPR17 plays an essential role in this regulation. GPR17 is highly expressed in the ventral progenitor regions of the neural tube and acts as a negative regulator of the Shh signal in chick embryos. Although the activation of the GPR17-related signal inhibits ventral identity, perturbation of Gpr17 expression leads to aberrant expansion of ventral neural domains. Notably, perturbation of Gpr17 expression partially inhibits the negative feedback of Gli activity. Moreover, GPR17 increases cAMP activity, suggesting that it exerts its function by inhibiting the processing of Gli3 protein. GPR17 also negatively regulates Shh signalling in neural cells differentiated from mouse embryonic stem cells, suggesting that GPR17 function is conserved among different organisms. Our results demonstrate that GPR17 is a novel negative regulator of Shh signalling in a wide range of cellular contexts.


Subject(s)
Adaptation, Physiological/physiology , Hedgehog Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Tube/embryology , Receptors, G-Protein-Coupled/metabolism , Animals , Body Patterning/physiology , Cell Differentiation/genetics , Chick Embryo , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryonic Development/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Transfection , Zinc Finger Protein Gli3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL