Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 118(5): 1262-1275, 2022 03 25.
Article in English | MEDLINE | ID: mdl-33909875

ABSTRACT

AIMS: Hepatic capillaries are lined with specialized liver sinusoidal endothelial cells (LSECs) which support macromolecule passage to hepatocytes and prevent fibrosis by keeping hepatic stellate cells (HSCs) quiescent. LSEC specialization is co-determined by transcription factors. The zinc-finger E-box-binding homeobox (Zeb)2 transcription factor is enriched in LSECs. Here, we aimed to elucidate the endothelium-specific role of Zeb2 during maintenance of the liver and in liver fibrosis. METHODS AND RESULTS: To study the role of Zeb2 in liver endothelium we generated EC-specific Zeb2 knock-out (ECKO) mice. Sequencing of liver EC RNA revealed that deficiency of Zeb2 results in prominent expression changes in angiogenesis-related genes. Accordingly, the vascular area was expanded and the presence of pillars inside ECKO liver vessels indicated that this was likely due to increased intussusceptive angiogenesis. LSEC marker expression was not profoundly affected and fenestrations were preserved upon Zeb2 deficiency. However, an increase in continuous EC markers suggested that Zeb2-deficient LSECs are more prone to dedifferentiation, a process called 'capillarization'. Changes in the endothelial expression of ligands that may be involved in HSC quiescence together with significant changes in the expression profile of HSCs showed that Zeb2 regulates LSEC-HSC communication and HSC activation. Accordingly, upon exposure to the hepatotoxin carbon tetrachloride (CCl4), livers of ECKO mice showed increased capillarization, HSC activation, and fibrosis compared to livers from wild-type littermates. The vascular maintenance and anti-fibrotic role of endothelial Zeb2 was confirmed in mice with EC-specific overexpression of Zeb2, as the latter resulted in reduced vascularity and attenuated CCl4-induced liver fibrosis. CONCLUSION: Endothelial Zeb2 preserves liver angioarchitecture and protects against liver fibrosis. Zeb2 and Zeb2-dependent genes in liver ECs may be exploited to design novel therapeutic strategies to attenuate hepatic fibrosis.


Subject(s)
Endothelial Cells , Liver Cirrhosis , Animals , Biomarkers/metabolism , Endothelial Cells/metabolism , Endothelium , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/prevention & control , Mice
2.
Brain Res ; 1749: 147140, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33022214

ABSTRACT

Neurodevelopmental disorders, including intellectual disability and autism spectrum disorder, are often caused by de novo autosomal dominant mutations. While mouse models are frequently used to investigate these disorders, the genetic background sometimes affects the appearance or severity of mutant phenotypes. In a previous report, we developed a system to produce de novo heterozygous mutant mice using the Cre-LoxP system without the need to maintain the heterozygous mutant line itself (Takagi et al. 2015). To further verify the applicability of the de novo mutation system in sperm, we used this system to produce a mouse model for Rubinstein-Taybi syndrome, using a Cbp heterozygous mutant, which has been reported to be difficult to maintain on a C57BL/6 background. Here, we show that de novo Cbp- loss-of-function heterozygous mutant mice with a C57BL/6 background, present with a clear craniofacial phenotype and reduced locomotor activity in the open field test, which was not observed in the loss-of-function of Cbp heterozygous mutant line mice with a mixed genetic background, but was observed in the dominant negative Cbp heterozygous mutant line with a mixed genetic background. Meanwhile, the de novo heterozygous Cbp mutant mice still showed great variability in survival rates despite their inbred background. These results further confirmed that the de novo mutation system used in germ cells is effective for stable production and analysis of an autosomal dominant disorder mouse model, which is often difficult to maintain as a mutant mouse line.


Subject(s)
CREB-Binding Protein/genetics , Disease Models, Animal , Mutation , Rubinstein-Taybi Syndrome/genetics , Spermatozoa/metabolism , Animals , CREB-Binding Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Rubinstein-Taybi Syndrome/metabolism
3.
Cereb Cortex ; 29(9): 3738-3751, 2019 08 14.
Article in English | MEDLINE | ID: mdl-30307479

ABSTRACT

SAD kinases regulate presynaptic vesicle clustering and neuronal polarization. A previous report demonstrated that Sada-/- and Sadb-/- double-mutant mice showed perinatal lethality with a severe defect in axon/dendrite differentiation, but their single mutants did not. These results indicated that they were functionally redundant. Surprisingly, we show that on a C57BL/6N background, SAD-A is essential for cortical development whereas SAD-B is dispensable. Sada-/- mice died within a few days after birth. Their cortical lamination pattern was disorganized and radial migration of cortical neurons was perturbed. Birth date analyses with BrdU and in utero electroporation using pCAG-EGFP vector showed a delayed migration of cortical neurons to the pial surface in Sada-/- mice. Time-lapse imaging of these mice confirmed slow migration velocity in the cortical plate. While the neurites of hippocampal neurons in Sada-/- mice could ultimately differentiate in culture to form axons and dendrites, the average length of their axons was shorter than that of the wild type. Thus, analysis on a different genetic background than that used initially revealed a nonredundant role for SAD-A in neuronal migration and differentiation.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/embryology , Cerebral Cortex/enzymology , Neurons/enzymology , Protein Serine-Threonine Kinases/physiology , Animals , Axons/enzymology , Cells, Cultured , Female , Isoenzymes , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
4.
Sci Rep ; 7: 46565, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28422173

ABSTRACT

Mowat-Wilson syndrome (MOWS) is a congenital disease caused by de novo heterozygous loss of function mutations or deletions of the ZEB2 gene. MOWS patients show multiple anomalies including intellectual disability, a distinctive facial appearance, microcephaly, congenital heart defects and Hirschsprung disease. However, the skin manifestation(s) of patients with MOWS has not been documented in detail. Here, we recognized that MOWS patients exhibit many Ehlers-Danlos syndrome (EDS)-like symptoms, such as skin hyperextensibility, atrophic scars and joint hypermobility. MOWS patients showed a thinner dermal thickness and electron microscopy revealed miniaturized collagen fibrils. Notably, mice with a mesoderm-specific deletion of the Zeb2 gene (Zeb2-cKO) demonstrated redundant skin, dermal hypoplasia and miniaturized collagen fibrils similar to those of MOWS patients. Dermal fibroblasts derived from Zeb2-cKO mice showed a decreased expression of extracellular matrix (ECM) molecules, such as collagens, whereas molecules involved in degradation of the ECM, such as matrix metalloproteinases (MMPs), were up-regulated. Furthermore, bleomycin-induced skin fibrosis was attenuated in Zeb2-cKO mice. We conclude that MOWS patients exhibit an EDS-like skin phenotype through alterations of collagen fibrillogenesis due to ZEB2 mutations or deletions.


Subject(s)
Collagen , Dermis , Ehlers-Danlos Syndrome , Facies , Hirschsprung Disease , Intellectual Disability , Microcephaly , Zinc Finger E-box Binding Homeobox 2 , Animals , Child , Child, Preschool , Collagen/genetics , Collagen/metabolism , Dermis/metabolism , Dermis/pathology , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/metabolism , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Knockout , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Proteolysis , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
5.
Sci Rep ; 7: 44111, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281676

ABSTRACT

Increasing evidence has suggested that human umbilical cord blood cells (hUCBC) have a favorable effect on hypoxic-ischemic (HI) brain injury. However, the efficacy of using hUCBCs to treat this injury has been variable and the underlying mechanism remains elusive. Here, we investigated its effectiveness using stereological analysis in an allogeneic system to examine whether intraperitoneal injection of cells derived from UCBCs of green fluorescent protein (GFP)-transgenic rats could ameliorate brain injury in neonatal rats. Three weeks after the HI event, the estimated residual brain volume was larger and motor function improved more in the cell-injected rats than in the control (PBS-treated) rats. The GFP-positive cells were hardly detectable in the brain (0.0057% of injected cells) 9 days after injection. Although 60% of GFP-positive cells in the brain were Iba1-positive, none of these were positive for NeuroD or DCX. While the number of proliferating cells increased in the hippocampus, that of activated microglia/macrophages decreased and a proportion of M2 microglia/macrophages increased in the ipsilateral hemisphere of cell-injected rats. These results suggest that intraperitoneal injection of cells derived from UCBCs could ameliorate HI injury, possibly through an endogenous response and not by supplying differentiated neurons derived from the injected stem cells.


Subject(s)
Fetal Blood/transplantation , Hypoxia-Ischemia, Brain/therapy , Animals , Animals, Newborn , Behavior, Animal , Cord Blood Stem Cell Transplantation/methods , Doublecortin Protein , Hippocampus/pathology , Hypoxia-Ischemia, Brain/complications , Inflammation/complications , Inflammation/prevention & control , Injections, Intraperitoneal , Motor Activity , Neurons/physiology , Rats , Rats, Transgenic , Transplantation, Homologous
6.
Proc Natl Acad Sci U S A ; 113(51): 14775-14780, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930303

ABSTRACT

Dendritic cells (DCs) and monocytes develop from a series of bone-marrow-resident progenitors in which lineage potential is regulated by distinct transcription factors. Zeb2 is an E-box-binding protein associated with epithelial-mesenchymal transition and is widely expressed among hematopoietic lineages. Previously, we observed that Zeb2 expression is differentially regulated in progenitors committed to classical DC (cDC) subsets in vivo. Using systems for inducible gene deletion, we uncover a requirement for Zeb2 in the development of Ly-6Chi monocytes but not neutrophils, and we show a corresponding requirement for Zeb2 in expression of the M-CSF receptor in the bone marrow. In addition, we confirm a requirement for Zeb2 in development of plasmacytoid DCs but find that Zeb2 is not required for cDC2 development. Instead, Zeb2 may act to repress cDC1 progenitor specification in the context of inflammatory signals.


Subject(s)
Dendritic Cells/cytology , Gene Expression Regulation , Monocytes/cytology , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/physiology , Animals , Bone Marrow/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Lineage , Cytoplasm/metabolism , Female , Flow Cytometry , Gene Deletion , Gene Expression Profiling , Inflammation , Integrases/metabolism , Male , Mice , Neutrophils/cytology , Neutrophils/metabolism
7.
Cell Rep ; 17(6): 1473-1481, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27806288

ABSTRACT

During spinal cord development, astrocyte precursors arise from neuroepithelial progenitors, delaminate from the ventricular zone, and migrate toward their final locations where they differentiate. Although the mechanisms underlying their early specification and late differentiation are being deciphered, less is known about the temporal control of their migration. Here, we show that the epithelial-mesenchymal transition regulator Zeb1 is expressed in glial precursors and report that loss of Zeb1 function specifically delays the onset of astrocyte precursor delamination from the ventricular zone, correlating with transient deregulation of the adhesion protein Cadherin-1. Consequently, astrocyte precursor invasion into the Zeb1-/- mutant white matter is delayed, and induction of their differentiation is postponed. These findings illustrate how fine regulation of adhesive properties influences the onset of neural precursor migration and further support the notion that duration of exposure of migrating astrocyte precursors to environmental cues and/or their correct positioning influence the timing of their differentiation.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cell Movement , Spinal Cord/cytology , Stem Cells/cytology , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Aging/genetics , Animals , Body Patterning , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Mutation/genetics
8.
Kidney Int ; 90(6): 1262-1273, 2016 12.
Article in English | MEDLINE | ID: mdl-27591083

ABSTRACT

Primary glomerulocystic kidney disease is a special form of renal cystic disorder characterized by Bowman's space dilatation in the absence of tubular cysts. ZEB2 is a SMAD-interacting transcription factor involved in Mowat-Wilson syndrome, a congenital disorder with an increased risk for kidney anomalies. Here we show that deletion of Zeb2 in mesenchyme-derived nephrons with either Pax2-cre or Six2-cre causes primary glomerulocystic kidney disease without tubular cysts in mice. Glomerulotubular junction analysis revealed many atubular glomeruli in the kidneys of Zeb2 knockout mice, which explains the presence of glomerular cysts in the absence of tubular dilatation. Gene expression analysis showed decreased expression of early proximal tubular markers in the kidneys of Zeb2 knockout mice preceding glomerular cyst formation, suggesting that defects in proximal tubule development during early nephrogenesis contribute to the formation of congenital atubular glomeruli. At the molecular level, Zeb2 deletion caused aberrant expression of Pkd1, Hnf1ß, and Glis3, three genes causing glomerular cysts. Thus, Zeb2 regulates the morphogenesis of mesenchyme-derived nephrons and is required for proximal tubule development and glomerulotubular junction formation. Our findings also suggest that ZEB2 might be a novel disease gene in patients with primary glomerular cystic disease.


Subject(s)
Central Nervous System Diseases/genetics , Dental Enamel/abnormalities , Diabetes Mellitus, Type 2/genetics , Homeodomain Proteins/physiology , Kidney Diseases, Cystic/genetics , Kidney/embryology , Repressor Proteins/physiology , Animals , DNA-Binding Proteins , Hepatocyte Nuclear Factor 1-beta/metabolism , Kidney/metabolism , Mice, Knockout , Repressor Proteins/metabolism , TRPP Cation Channels/metabolism , Trans-Activators/metabolism , Zinc Finger E-box Binding Homeobox 2
9.
Development ; 143(15): 2829-41, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27385012

ABSTRACT

The transcription factor Sip1 (Zeb2) plays multiple roles during CNS development from early acquisition of neural fate to cortical neurogenesis and gliogenesis. In humans, SIP1 (ZEB2) haploinsufficiency leads to Mowat-Wilson syndrome, a complex congenital anomaly including intellectual disability, epilepsy and Hirschsprung disease. Here we uncover the role of Sip1 in retinogenesis. Somatic deletion of Sip1 from mouse retinal progenitors primarily affects the generation of inner nuclear layer cell types, resulting in complete loss of horizontal cells and reduced numbers of amacrine and bipolar cells, while the number of Muller glia is increased. Molecular analysis places Sip1 downstream of the eye field transcription factor Pax6 and upstream of Ptf1a in the gene network required for generating the horizontal and amacrine lineages. Intriguingly, characterization of differentiation dynamics reveals that Sip1 has a role in promoting the timely differentiation of retinal interneurons, assuring generation of the proper number of the diverse neuronal and glial cell subtypes that constitute the functional retina in mammals.


Subject(s)
Nerve Tissue Proteins/metabolism , Retina/cytology , Retina/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Chromatin Immunoprecipitation , Female , Fluorescent Antibody Technique , Mice , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Neurogenesis/physiology , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Pregnancy , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Dev Growth Differ ; 58(2): 205-14, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26691438

ABSTRACT

The vertebrate Zfhx1 transcription factor family comprises δEF1 and Sip1, which bind to CACCT-containing sequences and act as transcriptional repressors. It has been a longstanding question whether these transcription factors share the same regulatory functions in vivo. It has been shown that neural crest (NC) delamination depends on the Sip1 activity at the cranial level in mouse and chicken embryos, and it remained unclear how NC delamination is regulated at the trunk level. We observed that the expression of δEF1 and Sip1 overlaps in many tissues in chicken embryos, including NC cells at the trunk level. To clarify the above questions, we separately knocked down δEF1 and Sip1 or in combination in NC cells by electroporation of vectors expressing short hairpin RNAs (shRNAs) against respective mRNAs on the dorsal side of neural tubes that generate NC cells. In all cases, the migrating NC cell population was significantly reduced, paralleled by the decreased expression of δEF1 or Sip1 targeted by shRNAs. Expression of Sox10, the major transcription factor that regulates NC development, was also decreased by the shRNAs against δEF1 or Sip1. We conclude that the trunk NC delamination is regulated by both δEF1 and Sip1 in an analogous manner, and that these transcription factors can share equivalent regulatory functions in embryonic tissues.


Subject(s)
Avian Proteins/metabolism , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Crest/embryology , Transcription Factors/metabolism , Animals , Chick Embryo , Mice , Neural Crest/cytology
11.
Mutagenesis ; 31(1): 61-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26220009

ABSTRACT

Cleidocranial dysplasia (CCD; MIM 119600) is an autosomal dominant skeletal dysplasia characterised by hypopalstic and/or aplastic clavicles, midface hypoplasia, absent or delayed closure of cranial sutures, moderately short stature, delayed eruption of permanent dentition and supernumerary teeth. The molecular pathogenesis can be explained in about two-thirds of CCD patients by haploinsufficiency of the RUNX2 gene. In our current study, we identified a novel and rare variant of the RUNX2 gene (c.181_189dupGCGGCGGCT) in a Japanese patient with phenotypic features of CCD. The insertion led an alanine tripeptide expansion (+3Ala) in the polyalanine tract. To date, a RUNX2 variant with alanine decapeptide expansion (+10Ala) is the only example of a causative variant of RUNX2 with polyalanine tract expansion to be reported, whilst RUNX2 (+1Ala) has been isolated from the healthy population. Thus, precise analyses of the RUNX2 (+3Ala) variant were needed to clarify whether the tripeptide expanded RUNX2 is a second disease-causing mutant with alanine tract expansion. We therefore investigated the biochemical properties of the mutant RUNX2 (+3Ala), which contains 20 alanine residues in the polyalanine tract. When transfected in COS7 cells, RUNX2 (+3Ala) formed intracellular ubiquitinated aggregates after 24h, and exerted a dominant negative effect in vitro. At 24h after gene transfection, whereas slight reduction was observed in RUNX2 (+10Ala), all of these mutants significantly activated osteoblast-specific element-2, a cis-acting sequence in the promoter of the RUNX2 target gene osteocalcin. The aggregation growth of RUNX2 (+3Ala) was clearly lower and slower than that of RUNX2 (+10Ala). Furthermore, we investigated several other RUNX2 variants with various alanine tract lengths, and found that the threshold for aggregation may be RUNX2 (+3Ala). We conclude that RUNX2 (+3Ala) is the cause of CCD in our current case, and that the accumulation of intracellular aggregates in vitro is related to the length of the alanine tract.


Subject(s)
Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Trinucleotide Repeat Expansion , Adult , Asian People/genetics , Cell Line , Cleidocranial Dysplasia/diagnosis , Cleidocranial Dysplasia/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Japan , Osteocalcin/metabolism , Peptides , Transcriptional Activation
12.
J Exp Med ; 212(12): 2027-39, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26503445

ABSTRACT

ZEB2 is a multi-zinc-finger transcription factor known to play a significant role in early neurogenesis and in epithelial-mesenchymal transition-dependent tumor metastasis. Although the function of ZEB2 in T lymphocytes is unknown, activity of the closely related family member ZEB1 has been implicated in lymphocyte development. Here, we find that ZEB2 expression is up-regulated by activated T cells, specifically in the KLRG1(hi) effector CD8(+) T cell subset. Loss of ZEB2 expression results in a significant loss of antigen-specific CD8(+) T cells after primary and secondary infection with a severe impairment in the generation of the KLRG1(hi) effector memory cell population. We show that ZEB2, which can bind DNA at tandem, consensus E-box sites, regulates gene expression of several E-protein targets and may directly repress Il7r and Il2 in CD8(+) T cells responding to infection. Furthermore, we find that T-bet binds to highly conserved T-box sites in the Zeb2 gene and that T-bet and ZEB2 regulate similar gene expression programs in effector T cells, suggesting that T-bet acts upstream and through regulation of ZEB2. Collectively, we place ZEB2 in a larger transcriptional network that is responsible for the balance between terminal differentiation and formation of memory CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeodomain Proteins/immunology , Lymphocytic Choriomeningitis/immunology , Repressor Proteins/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Flow Cytometry , Homeodomain Proteins/genetics , Host-Pathogen Interactions/immunology , Humans , Immunologic Memory/immunology , Lectins, C-Type , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Protein Binding/immunology , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome/genetics , Transcriptome/immunology , Zinc Finger E-box Binding Homeobox 2
13.
Hum Mol Genet ; 24(22): 6390-402, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26319231

ABSTRACT

Mowat-Wilson syndrome (MOWS) is caused by de novo heterozygous mutation at ZEB2 (SIP1, ZFHX1B) gene, and exhibit moderate to severe intellectual disability (ID), a characteristic facial appearance, epilepsy and other congenital anomalies. Establishing a murine MOWS model is important, not only for investigating the pathogenesis of this disease, but also for identifying compounds that may improve the symptoms. However, because the heterozygous Zeb2 knockout mouse could not be maintained as a mouse line with the inbred C57BL/6 background, it was difficult to use those mice for the study of MOWS. Here, we systematically generated de novo Zeb2 Δex7/+ mice by inducing the Zeb2 mutation in the germ cells using conditional recombination system. The de novo Zeb2 Δex7/+ mice with C57BL/6 background developed multiple defects relevant to MOWS, including craniofacial abnormalities, defective corpus callosum formation and the decreased number of parvalbumin interneurons in the cortex. In behavioral analyses, these mice showed reduced motor activity, increased anxiety and impaired sociability. Notably, during the Barnes maze test, immobile Zeb2 mutant mice were observed over repeated trials. In contrast, neither the mouse line nor the de novo Zeb2 Δex7/+ mice with the closed colony ICR background showed cranial abnormalities or reduced motor activities. These results demonstrate the advantages of using de novo Zeb2 Δex7/+ mice with the C57BL/6 background as the MOWS model. To our knowledge, this is the first time an inducible de novo mutation system has been applied to murine germline cells to produce an animal model of a human congenital disease.


Subject(s)
Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intellectual Disability/genetics , Microcephaly/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Aicardi Syndrome/genetics , Aicardi Syndrome/metabolism , Animals , Cerebral Cortex/metabolism , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Disease Models, Animal , Epilepsy/genetics , Epilepsy/metabolism , Facies , Female , Genetic Association Studies , Germ Cells , Germ-Line Mutation , Heterozygote , Hirschsprung Disease/metabolism , Humans , Intellectual Disability/metabolism , Male , Mental Disorders/genetics , Mental Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Microcephaly/metabolism , Zinc Finger E-box Binding Homeobox 2
14.
PLoS One ; 10(6): e0128227, 2015.
Article in English | MEDLINE | ID: mdl-26030286

ABSTRACT

Congenital tooth agenesis is caused by mutations in the MSX1, PAX9, WNT10A, or AXIN2 genes. Here, we report a Japanese family with nonsyndromic tooth agenesis caused by a novel nucleotide substitution in the intronic region between exons 1 and 2 of the MSX1 gene. Because the mutation is located 9 bp before exon 2 (c.452-9G>A), we speculated that the nucleotide substitution would generate an abnormal splice site. Using cDNA analysis of an immortalized patient blood cell, we confirmed that an additional 7-nucleotide sequence was inserted at the splice junction between exons 1 and 2 (c.451_452insCCCTCAG). The consequent frameshift generated a homeodomain-truncated MSX1 (p.R151fsX20). We then studied the subcellular localization of truncated MSX1 protein in COS cells, and observed that it had a whole cell distribution more than a nuclear localization, compared to that of wild-type protein. This result suggests a deletion of the nuclear localization signal, which is mapped to the MSX1 homeodomain. These results indicate that this novel intronic nucleotide substitution is the cause of tooth agenesis in this family. To date, most MSX1 variants isolated from patients with tooth agenesis involve single amino acid substitutions in the highly conserved homeodomain or deletion mutants caused by frameshift or nonsense mutations. We here report a rare case of an intronic mutation of the MSX1 gene responsible for human tooth agenesis. In addition, the missing tooth patterns were slightly but significantly different between an affected monozygotic twin pair of this family, showing that epigenetic or environmental factors also affect the phenotypic variations of missing teeth among patients with nonsyndromic tooth agenesis caused by an MSX1 haploinsufficiency.


Subject(s)
Anodontia/genetics , Asian People/genetics , Introns/genetics , MSX1 Transcription Factor/genetics , Nucleotides/genetics , RNA Splice Sites/genetics , Adult , Anodontia/diagnostic imaging , Base Sequence , Blotting, Western , DNA Mutational Analysis , DNA, Complementary/genetics , Family , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Pedigree , RNA Splicing/genetics , Radiography , Subcellular Fractions/metabolism
15.
PLoS One ; 9(8): e102944, 2014.
Article in English | MEDLINE | ID: mdl-25101640

ABSTRACT

Since MSX1 and PAX9 are linked to the pathogenesis of nonsyndromic tooth agenesis, we performed detailed mutational analysis of these two genes sampled from Japanese patients. We identified two novel MSX1 variants with an amino acid substitution within the homeodomain; Thr174Ile (T174I) from a sporadic hypodontia case and Leu205Arg (L205R) from a familial oligodontia case. Both the Thr174 and Leu205 residues in the MSX1 homeodomain are highly conserved among different species. To define possible roles of mutations at these amino acids in the pathogenesis of nonsyndromic tooth agenesis, we performed several functional analyses. It has been demonstrated that MSX1 plays a pivotal role in hard tissue development as a suppressor for mesenchymal cell differentiation. To evaluate the suppression activity of the variants in mesenchymal cells, we used the myoD-promoter, which is one of convenient reporter assay system for MSX1. Although the gene products of these MSX1 variants are stable and capable of normal nuclear localization, they do not suppress myoD-promoter activity in differentiated C2C12 cells. To clarify the molecular mechanisms underlying our results, we performed further analyses including electrophoretic mobility shift assays, and co-immunoprecipitation assays to survey the molecular interactions between the mutant MSX1 proteins and the oligonucleotide DNA with MSX1 consensus binding motif or EZH2 methyltransferase. Since EZH2 is reported to interact with MSX1 and regulate MSX1 mediated gene suppression, we hypothesized that the T174I and L205R substitutions would impair this interaction. We conclude from the results of our experiments that the DNA binding ability of MSX1 is abolished by these two amino acid substitutions. This illustrates a causative role of the T174I and L205R MSX1 homeodomain mutations in tooth agenesis, and suggests that they may influence cell proliferation and differentiation resulting in lesser tooth germ formation in vivo.


Subject(s)
Amino Acid Substitution , Anodontia/genetics , MSX1 Transcription Factor/genetics , Amino Acid Sequence , Binding Sites , Conserved Sequence , DNA Mutational Analysis , Electrophoretic Mobility Shift Assay , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation , Humans , Japan , Male , Molecular Sequence Data , Pedigree , Polycomb Repressive Complex 2/metabolism , Sequence Alignment , Sequence Analysis, Protein
16.
Eur J Oral Sci ; 122(1): 15-20, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24329876

ABSTRACT

Nonsyndromic tooth agenesis is one of the most common anomalies in human development. Part of the malformation is inherited and is associated with paired box 9 (PAX9), msh homeobox 1 (MSX1), and axin 2 (AXIN2) mutations. To obtain a comprehensive understanding of the genetic and molecular mechanisms that underlie this genetic disease, we investigated six familial and seven sporadic Japanese cases of nonsyndromic tooth agenesis. Searches for mutations in these candidate genes detected a novel nonsense mutation (c.416G>A) in exon 1 of MSX1 from a family with oligodontia. This mutation co-segregated in the affected family members. Moreover, this mutation produced a termination codon in the first exon and therefore the gene product (W139X) was truncated at the C terminus, hence, the entire homeodomain/MH4, which has many functions, such as DNA binding, protein-protein interaction, and nuclear localization, was absent. We characterized the properties of this truncated MSX1 by investigating the subcellular localization of the mutant gene product in transfected cells. The wild-type MSX1 localized exclusively at the nuclear periphery of transfected cells, whereas the mutant MSX1 was stable but localized diffusely throughout the whole cell. These results indicate that W139X MSX1 is responsible for tooth agenesis.


Subject(s)
Anodontia/genetics , Codon, Nonsense/genetics , MSX1 Transcription Factor/genetics , Adenine , Anodontia/pathology , Axin Protein/genetics , Cell Culture Techniques , Cell Nucleus/ultrastructure , Chromosome Segregation/genetics , Codon, Terminator/genetics , Dinucleotide Repeats/genetics , Exons/genetics , Female , Genes, Homeobox/genetics , Guanine , HEK293 Cells , Humans , Male , Middle Aged , PAX9 Transcription Factor/genetics , Tryptophan/genetics , Young Adult
17.
Genesis ; 52(1): 56-67, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24243579

ABSTRACT

A loss of function of SIP1 (Smad interacting protein 1) in the mouse as well as in human of Mowat-Wilson syndrome results in severe and multiple defects in neural tissue development, especially in the brain. However, no detailed expression analysis of SIP1 during brain development has been previously reported. In this study, we describe the generation of an EGFP knock-in reporter mouse for the Sip1 locus and our subsequent analysis of SIP1-EGFP fusion protein expression during brain development. SIP1-EGFP expression was observed in the pyramidal neurons of the hippocampus, the dentate gyrus, and the postmitotic neurons in the cerebral cortex. In layer 5 of the cerebral cortex, SIP1-EGFP expression was complementary to the Ctip2-expressing neurons, most of which are thought to be the cortico-spinal neurons. This suggested that SIP1-EGFP expressing cells might have the specific trajectory targets other than the spinal region. We further observed SIP1-EGFP expression in oligodendrocytes of the corpus callosum and fimbria, Bergmann glial cells of the cerebellum, the olfactory bulb, and in the serotonergic and dopaminergic neurons of the raphe nuclei in the brainstem. These findings may help to clarify the unknown roles of SIP1 in these cells and the pathoetiology of Mowat-Wilson syndrome.


Subject(s)
Brain/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Brain/growth & development , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Corpus Callosum/metabolism , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Facies , Gene Knock-In Techniques , Genes, Reporter , Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Humans , Intellectual Disability/genetics , Mice , Mice, Inbred C57BL , Microcephaly/genetics , Pyramidal Cells/metabolism , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics , Zinc Finger E-box Binding Homeobox 2
18.
Neuron ; 77(1): 83-98, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23312518

ABSTRACT

Mammalian pallial (cortical and hippocampal) and striatal interneurons are both generated in the embryonic subpallium, including the medial ganglionic eminence (MGE). Herein we demonstrate that the Zfhx1b (Sip1, Zeb2) zinc finger homeobox gene is required in the MGE, directly downstream of Dlx1&2, to generate cortical interneurons that express Cxcr7, MafB, and cMaf. In its absence, Nkx2-1 expression is not repressed, and cells that ordinarily would become cortical interneurons appear to transform toward a subtype of GABAergic striatal interneurons. These results show that Zfhx1b is required to generate cortical interneurons, and suggest a mechanism for the epilepsy observed in humans with Zfhx1b mutations (Mowat-Wilson syndrome).


Subject(s)
Cerebral Cortex/embryology , Corpus Striatum/embryology , Homeodomain Proteins/biosynthesis , Interneurons/physiology , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Animals , Animals, Newborn , Base Sequence , Cells, Cultured , Cerebral Cortex/growth & development , Corpus Striatum/growth & development , Gene Expression Regulation, Developmental , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Neurogenesis/physiology , Repressor Proteins/genetics , Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2
19.
Ocul Surf ; 10(4): 217-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23084142

ABSTRACT

Epithelial-mesenchymal interactions and epithelial-to-mesenchymal transition (EMT) are essential during tissue formation and organ morphogenesis. The roles of Wnt/ß-catenin signaling have been studied in many organ systems. In this review, we describe the importance of Wnt/ß-catenin signaling by comparing skin and corneal development of Wnt/ß-catenin gain of function (GOF) mutant mice. In the skin, Wnt/ß-catenin signals have been suggested to play essential roles in regulating cell-cell interaction, cell proliferation and differentiation. Wnt signaling may be associated with basal cell carcinoma (BCC) of the skin. In the case of cornea, ß-catenin GOF mutation leads to epithelial hyperplasia. Investigation of the mechanisms of growth factor signaling as a reference to general organogenesis could provide profound insights for understanding corneal development and pathogenesis.


Subject(s)
Cornea/embryology , Cornea/metabolism , Epithelium/physiology , Mesoderm/physiology , Signal Transduction/physiology , Animals , Mice , Mice, Mutant Strains , Skin/embryology , Skin/metabolism
20.
Neuron ; 73(4): 713-28, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22365546

ABSTRACT

Myelination by oligodendrocytes in the central nervous system (CNS) is essential for proper brain function, yet the molecular determinants that control this process remain poorly understood. The basic helix-loop-helix transcription factors Olig1 and Olig2 promote myelination, whereas bone morphogenetic protein (BMP) and Wnt/ß-catenin signaling inhibit myelination. Here we show that these opposing regulators of myelination are functionally linked by the Olig1/2 common target Smad-interacting protein-1 (Sip1). We demonstrate that Sip1 is an essential modulator of CNS myelination. Sip1 represses differentiation inhibitory signals by antagonizing BMP receptor-activated Smad activity while activating crucial oligodendrocyte-promoting factors. Importantly, a key Sip1-activated target, Smad7, is required for oligodendrocyte differentiation and partially rescues differentiation defects caused by Sip1 loss. Smad7 promotes myelination by blocking the BMP- and ß-catenin-negative regulatory pathways. Thus, our findings reveal that Sip1-mediated antagonism of inhibitory signaling is critical for promoting CNS myelination and point to new mediators for myelin repair.


Subject(s)
Central Nervous System/physiology , Gene Expression Regulation, Developmental/genetics , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Smad Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Proteins/metabolism , Caspase 3/metabolism , Cell Differentiation/genetics , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/ultrastructure , Embryo, Mammalian , Facies , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Homeodomain Proteins/metabolism , Humans , Immunoprecipitation , Intellectual Disability/genetics , Intellectual Disability/pathology , Ki-67 Antigen/metabolism , Mice , Mice, Knockout , Microcephaly/genetics , Microcephaly/pathology , Microscopy, Electron, Transmission , Models, Molecular , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Oligonucleotide Array Sequence Analysis , Optic Nerve/embryology , Optic Nerve/growth & development , Optic Nerve/metabolism , Organogenesis , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Smad Proteins/genetics , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transfection , Zinc Finger E-box Binding Homeobox 2
SELECTION OF CITATIONS
SEARCH DETAIL
...