Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Open Biol ; 14(6): 240113, 2024 Jun.
Article En | MEDLINE | ID: mdl-38889770

Autism spectrum disorder (ASD) is a group of neurodevelopmental conditions associated with deficits in social interaction and communication, together with repetitive behaviours. The cell adhesion molecule protocadherin10 (PCDH10) is linked to ASD in humans. Pcdh10 is expressed in the nervous system during embryonic and early postnatal development and is important for neural circuit formation. In mice, strong expression of Pcdh10 in the ganglionic eminences and in the basolateral complex (BLC) of the amygdala was observed at mid and late embryonic stages, respectively. Both inhibitory and excitatory neurons expressed Pcdh10 in the BLC at perinatal stages and vocalization-related genes were enriched in Pcdh10-expressing neurons in adult mice. An epitope-tagged Pcdh10-HAV5 mouse line revealed endogenous interactions of PCDH10 with synaptic proteins in the young postnatal telencephalon. Nuanced socio-affective communication changes in call emission rates, acoustic features and call subtype clustering were primarily observed in heterozygous pups of a conditional knockout (cKO) with selective deletion of Pcdh10 in Gsh2-lineage interneurons. These changes were less prominent in heterozygous ubiquitous Pcdh10 KO pups, suggesting that altered anxiety levels associated with Gsh2-lineage interneuron functioning might drive the behavioural effects. Together, loss of Pcdh10 specifically in interneurons contributes to behavioural alterations in socio-affective communication with relevance to ASD.


Amygdala , Cadherins , Interneurons , Mice, Knockout , Protocadherins , Animals , Cadherins/metabolism , Cadherins/genetics , Interneurons/metabolism , Mice , Protocadherins/metabolism , Amygdala/metabolism , Amygdala/growth & development , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Vocalization, Animal/physiology , Male , Social Behavior
2.
Nat Commun ; 14(1): 1210, 2023 03 03.
Article En | MEDLINE | ID: mdl-36869101

Early during preimplantation development and in heterogeneous mouse embryonic stem cells (mESC) culture, pluripotent cells are specified towards either the primed epiblast or the primitive endoderm (PE) lineage. Canonical Wnt signaling is crucial for safeguarding naive pluripotency and embryo implantation, yet the role and relevance of canonical Wnt inhibition during early mammalian development remains unknown. Here, we demonstrate that transcriptional repression exerted by Wnt/TCF7L1 promotes PE differentiation of mESCs and in preimplantation inner cell mass. Time-series RNA sequencing and promoter occupancy data reveal that TCF7L1 binds and represses genes encoding essential naive pluripotency factors and indispensable regulators of the formative pluripotency program, including Otx2 and Lef1. Consequently, TCF7L1 promotes pluripotency exit and suppresses epiblast lineage formation, thereby driving cells into PE specification. Conversely, TCF7L1 is required for PE specification as deletion of Tcf7l1 abrogates PE differentiation without restraining epiblast priming. Taken together, our study underscores the importance of transcriptional Wnt inhibition in regulating lineage specification in ESCs and preimplantation embryo development as well as identifies TCF7L1 as key regulator of this process.


Automobile Driving , Endoderm , Transcription Factor 7-Like 1 Protein , Animals , Female , Mice , Pregnancy , Blastocyst , Cell Differentiation , Germ Layers
3.
Development ; 147(10)2020 05 26.
Article En | MEDLINE | ID: mdl-32253238

The transcription factor Zeb2 controls fate specification and subsequent differentiation and maturation of multiple cell types in various embryonic tissues. It binds many protein partners, including activated Smad proteins and the NuRD co-repressor complex. How Zeb2 subdomains support cell differentiation in various contexts has remained elusive. Here, we studied the role of Zeb2 and its domains in neurogenesis and neural differentiation in the young postnatal ventricular-subventricular zone (V-SVZ), in which neural stem cells generate olfactory bulb-destined interneurons. Conditional Zeb2 knockouts and separate acute loss- and gain-of-function approaches indicated that Zeb2 is essential for controlling apoptosis and neuronal differentiation of V-SVZ progenitors before and after birth, and we identified Sox6 as a potential downstream target gene of Zeb2. Zeb2 genetic inactivation impaired the differentiation potential of the V-SVZ niche in a cell-autonomous fashion. We also provide evidence that its normal function in the V-SVZ also involves non-autonomous mechanisms. Additionally, we demonstrate distinct roles for Zeb2 protein-binding domains, suggesting that Zeb2 partners co-determine neuronal output from the mouse V-SVZ in both quantitative and qualitative ways in early postnatal life.


Lateral Ventricles/embryology , Lateral Ventricles/growth & development , Neurogenesis/genetics , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Knockout Techniques , Interneurons/metabolism , Lateral Ventricles/metabolism , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Olfactory Bulb/metabolism , SOXD Transcription Factors/metabolism , Signal Transduction/immunology , Zinc Finger E-box Binding Homeobox 2/genetics
5.
Stem Cells ; 35(3): 611-625, 2017 03.
Article En | MEDLINE | ID: mdl-27739137

In human embryonic stem cells (ESCs) the transcription factor Zeb2 regulates neuroectoderm versus mesendoderm formation, but it is unclear how Zeb2 affects the global transcriptional regulatory network in these cell-fate decisions. We generated Zeb2 knockout (KO) mouse ESCs, subjected them as embryoid bodies (EBs) to neural and general differentiation and carried out temporal RNA-sequencing (RNA-seq) and reduced representation bisulfite sequencing (RRBS) analysis in neural differentiation. This shows that Zeb2 acts preferentially as a transcriptional repressor associated with developmental progression and that Zeb2 KO ESCs can exit from their naïve state. However, most cells in these EBs stall in an early epiblast-like state and are impaired in both neural and mesendodermal differentiation. Genes involved in pluripotency, epithelial-to-mesenchymal transition (EMT), and DNA-(de)methylation, including Tet1, are deregulated in the absence of Zeb2. The observed elevated Tet1 levels in the mutant cells and the knowledge of previously mapped Tet1-binding sites correlate with loss-of-methylation in neural-stimulating conditions, however, after the cells initially acquired the correct DNA-methyl marks. Interestingly, cells from such Zeb2 KO EBs maintain the ability to re-adapt to 2i + LIF conditions even after prolonged differentiation, while knockdown of Tet1 partially rescues their impaired differentiation. Hence, in addition to its role in EMT, Zeb2 is critical in ESCs for exit from the epiblast state, and links the pluripotency network and DNA-methylation with irreversible commitment to differentiation. Stem Cells 2017;35:611-625.


Cell Lineage , Germ Layers/cytology , Germ Layers/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Cell Differentiation , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Mice , Mice, Knockout , Neurons/cytology , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Principal Component Analysis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Sequence Analysis, RNA , Transcription, Genetic
6.
PLoS One ; 10(5): e0124974, 2015.
Article En | MEDLINE | ID: mdl-25996609

Patient-derived tumor xenograft (PDTX) approach is nowadays considered a reliable preclinical model to study in vivo cancer biology and therapeutic response. NOD scid and Il2rg-deficient mice represent the "gold standard" host for the generation of PDTXs. Compared to other immunocompromised murine lines, these mice offers several advantages including higher engraftment rate, longer lifespan and improved morphological and molecular preservation of patient-derived neoplasms. Here we describe a spectrum of previously uncharacterized post-transplant disorders affecting 14/116 (12%) NOD.Cg- Prkdcscid Il2rgtm1Sug/JicTac (NOG) mice subcutaneously engrafted with patient-derived metastatic melanomas. Affected mice exhibited extensive scaling/crusting dermatitis (13/14) associated with emaciation (13/14) and poor/unsuccessful tumor engraftment (14/14). In this context, the following pathological conditions have been recognized and characterized in details: (i) immunoinflammatory disorders with features of graft versus host disease (14/14); (ii) reactive lymphoid infiltrates effacing xenografted tumors (8/14); (iii) post-transplant B cell lymphomas associated with Epstein-Barr virus reactivation (2/14). We demonstrate that all these entities are driven by co-transplanted human immune cells populating patient-derived tumor samples. Since the exploding interest in the utilization of NOD scid and Il2rg-deficient mice for the establishment of PDTX platforms, it is of uppermost importance to raise the awareness of the limitations associated with this model. The disorders here described adversely impact tumor engraftment rate and animal lifespan, potentially representing a major confounding factor in the context of efficacy and personalized therapy studies. The occurrence of these conditions in the NOG model reflects the ability of this mouse line to promote efficient engraftment of human immune cells. Co-transplanted human lymphoid cells have indeed the potential to colonize the recipient mouse initiating the post-transplant conditions here reported. On the other hand, the evidence of an immune response of human origin against the xenotransplanted melanoma opens intriguing perspectives for the establishment of suitable preclinical models of anti-melanoma immunotherapy.


Graft vs Host Disease/etiology , Melanoma/pathology , Animals , Biopsy , Disease Models, Animal , Female , Heterografts , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation/adverse effects
7.
Neuron ; 77(1): 70-82, 2013 Jan 09.
Article En | MEDLINE | ID: mdl-23312517

GABAergic interneurons mainly originate in the medial ganglionic eminence (MGE) of the embryonic ventral telencephalon (VT) and migrate tangentially to the cortex, guided by membrane-bound and secreted factors. We found that Sip1 (Zfhx1b, Zeb2), a transcription factor enriched in migrating cortical interneurons, is required for their proper differentiation and correct guidance. The majority of Sip1 knockout interneurons fail to migrate to the neocortex and stall in the VT. RNA sequencing reveals that Sip1 knockout interneurons do not acquire a fully mature cortical interneuron identity and contain increased levels of the repulsive receptor Unc5b. Focal electroporation of Unc5b-encoding vectors in the MGE of wild-type brain slices disturbs migration to the neocortex, whereas reducing Unc5b levels in Sip1 knockout slices and brains rescues the migration defect. Our results reveal that Sip1, through tuning of Unc5b levels, is essential for cortical interneuron guidance.


Cell Movement/physiology , Cerebral Cortex/growth & development , Interneurons/physiology , Neocortex/growth & development , Nerve Tissue Proteins/deficiency , Receptors, Cell Surface/deficiency , Animals , Cerebral Cortex/cytology , Gene Knockout Techniques , Mice , Mice, Transgenic , Neocortex/cytology , Nerve Tissue Proteins/genetics , Netrin Receptors , Organ Culture Techniques , Receptors, Cell Surface/genetics , Telencephalon/cytology , Telencephalon/growth & development
8.
Dev Cell ; 22(3): 501-14, 2012 Mar 13.
Article En | MEDLINE | ID: mdl-22364862

Gradients of vascular endothelial growth factor (VEGF) induce single endothelial cells to become leading tip cells of emerging angiogenic sprouts. Tip cells then suppress tip-cell features in adjacent stalk cells via Dll4/Notch-mediated lateral inhibition. We report here that Smad1/Smad5-mediated BMP signaling synergizes with Notch signaling during selection of tip and stalk cells. Endothelium-specific inactivation of Smad1/Smad5 in mouse embryos results in impaired Dll4/Notch signaling and increased numbers of tip-cell-like cells at the expense of stalk cells. Smad1/5 downregulation in cultured endothelial cells reduced the expression of several target genes of Notch and of other stalk-cell-enriched transcripts (Hes1, Hey1, Jagged1, VEGFR1, and Id1-3). Moreover, Id proteins act as competence factors for stalk cells and form complexes with Hes1, which augment Hes1 levels in the endothelium. Our findings provide in vivo evidence for a regulatory loop between BMP/TGFß-Smad1/5 and Notch signaling that orchestrates tip- versus stalk-cell selection and vessel plasticity.


Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Adaptor Proteins, Signal Transducing , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Calcium-Binding Proteins/biosynthesis , Cell Cycle Proteins/biosynthesis , Cells, Cultured , Down-Regulation , Homeodomain Proteins/biosynthesis , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic , Phenotype , Serrate-Jagged Proteins , Smad1 Protein/genetics , Smad5 Protein/genetics , Transcription Factor HES-1 , Vascular Endothelial Growth Factor Receptor-1/biosynthesis
9.
Cytokine Growth Factor Rev ; 22(5-6): 287-300, 2011.
Article En | MEDLINE | ID: mdl-22119658

Signaling by the many ligands of the TGFß family strongly converges towards only five receptor-activated, intracellular Smad proteins, which fall into two classes i.e. Smad2/3 and Smad1/5/8, respectively. These Smads bind to a surprisingly high number of Smad-interacting proteins (SIPs), many of which are transcription factors (TFs) that co-operate in Smad-controlled target gene transcription in a cell type and context specific manner. A combination of functional analyses in vivo as well as in cell cultures and biochemical studies has revealed the enormous versatility of the Smad proteins. Smads and their SIPs regulate diverse molecular and cellular processes and are also directly relevant to development and disease. In this survey, we selected appropriate examples on the BMP-Smads, with emphasis on Smad1 and Smad5, and on a number of SIPs, i.e. the CPSF subunit Smicl, Ttrap (Tdp2) and Sip1 (Zeb2, Zfhx1b) from our own research carried out in three different vertebrate models.


Bone Morphogenetic Proteins/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Signal Transduction
10.
Int J Dev Biol ; 53(7): 1045-51, 2009.
Article En | MEDLINE | ID: mdl-19598121

Gene trapping in mouse embryonic stem (ES) cells enables near-saturation vector-based insertional mutagenesis across the genome of this model organism. About 135,000 trapped ES cell lines are made available to the scientific community by the International Gene Trap Consortium (IGTC; www.genetrap.org). A search of one of its databases identified an ES cell line (RRS512) with a betaGeo-based gene trap (gt) vector insertion in intron 5 of Ttrap, a gene that encodes an intracellular signalling protein, which is implicated in gastrulation movement and left-right asymmetry in zebrafish embryos. We have determined the exact gt insertion point in the mutant ES cell clone RRS512 and confirmed the production of a chimaeric transcript consisting of the upstream Ttrap exons and the gene trap vector encoded marker/selection fusion sequences. This ES cell line was used to generate heterozygous Ttrap mutant mice, which were further crossed to obtain Ttrap(gt/gt) mice. In contrast to Ttraps documented essential role during nodal and Smad3 controlled zebrafish early embryogenesis, Ttrap(gt/gt) mice were born with a normal Mendelian distribution. However, subsequent analysis of these Ttrap(gt/gt) mice has revealed a duplication of the wild-type Ttrap allele that was already present in the RRS512 cell line. Based on our detailed analysis presented here, we suggest an extensive procedure for the characterization of gene trap ES cell lines prior to generating gene trap mice with these.


Embryonic Stem Cells/metabolism , Genetic Vectors , Mutagenesis, Insertional/methods , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Female , Genetic Techniques , In Situ Hybridization , Male , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
11.
BMJ Case Rep ; 20092009.
Article En | MEDLINE | ID: mdl-21720541

Kabuki syndrome (KS) is a rare, congenital mental retardation syndrome. The aetiology of KS remains unknown. Four carefully selected patients with KS were screened for chromosomal imbalances using array comparative genomic hybridisation at 1 Mb resolution. In one patient, a 250 kb de novo microdeletion at 20p12.1 was detected, deleting exon 5 of C20orf133. The function of this gene is unknown. In situ hybridisation with the mouse orthologue of C20orf133 showed expression mainly in brain. The de novo nature of the deletion, the expression data and the fact that C20orf133 carries a macro domain, suggesting a role for the gene in chromatin biology, make the gene a likely candidate to cause the phenotype in this patient with KS. Both the finding of different of chromosomal rearrangements in patients with KS features and the absence of C20orf133 mutations in 19 additional patients with KS suggest that KS is genetically heterogeneous.

12.
J Med Genet ; 44(9): 562-9, 2007 Sep.
Article En | MEDLINE | ID: mdl-17586838

BACKGROUND: Kabuki syndrome (KS) is a rare, clinically recognisable, congenital mental retardation syndrome. The aetiology of KS remains unknown. METHODS: Four carefully selected patients with KS were screened for chromosomal imbalances using array comparative genomic hybridisation at 1 Mb resolution. RESULTS: In one patient, a 250 kb de novo microdeletion at 20p12.1 was detected, deleting exon 5 of C20orf133. The function of this gene is unknown. In situ hybridisation with the mouse orthologue of C20orf133 showed expression mainly in brain, but also in kidney, eye, inner ear, ganglia of the peripheral nervous system and lung. CONCLUSION: The de novo nature of the deletion, the expression data and the fact that C20orf133 carries a macro domain, suggesting a role for the gene in chromatin biology, make the gene a likely candidate to cause the phenotype in this patient with KS. Both the finding of different of chromosomal rearrangements in patients with KS features and the absence of C20orf133 mutations in 19 additional patients with KS suggest that KS is genetically heterogeneous.


Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 20/genetics , Intellectual Disability/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 20/chemistry , Chromosomes, Human, Pair 20/ultrastructure , DNA Repair Enzymes , Exons/genetics , Face/abnormalities , Female , Gene Expression Regulation, Developmental , Humans , Hydrolases , Infant, Newborn , Membrane Glycoproteins , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Nucleic Acid Hybridization , Organ Specificity , Phenotype , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Syndrome , Transcription Factors/deficiency , Transcription Factors/physiology
13.
Hum Mol Genet ; 16(12): 1423-36, 2007 Jun 15.
Article En | MEDLINE | ID: mdl-17478475

Mowat-Wilson syndrome is a recently delineated autosomal dominant developmental anomaly, whereby heterozygous mutations in the ZFHX1B gene cause mental retardation, delayed motor development, epilepsy and a wide spectrum of clinically heterogeneous features, suggestive of neurocristopathies at the cephalic, cardiac and vagal levels. However, our understanding of the etiology of this condition at the cellular level remains vague. This study presents the Zfhx1b protein expression domain in mouse embryos and correlates this with a novel mouse model involving a conditional mutation in the Zfhx1b gene in neural crest precursor cells. These mutant mice display craniofacial and gastrointestinal malformations that show resemblance to those found in human patients with Mowat-Wilson syndrome. In addition to these clinically recognized alterations, we document developmental defects in the heart, melanoblasts and sympathetic and parasympathetic anlagen. The latter observations in our mouse model for Mowat-Wilson suggest a hitherto unknown role for Zfhx1b in the development of these particular neural crest derivatives, which is a set of observations that should be acknowledged in the clinical management of this genetic disorder.


Abnormalities, Multiple/genetics , Homeodomain Proteins/genetics , Neural Crest/embryology , Repressor Proteins/genetics , Abnormalities, Multiple/embryology , Animals , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Female , Ganglia, Sensory/embryology , Ganglia, Sensory/metabolism , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Male , Melanocytes/metabolism , Mice , Mice, Knockout , Mutation , Neural Crest/abnormalities , Neural Crest/metabolism , Repressor Proteins/metabolism , Skull/embryology , Skull/metabolism , Sympathetic Nervous System/embryology , Sympathetic Nervous System/metabolism , Syndrome , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Zinc Finger E-box Binding Homeobox 2
14.
Development ; 133(17): 3399-409, 2006 Sep.
Article En | MEDLINE | ID: mdl-16887830

Smad5 is an intracellular mediator of bone morphogenetic protein (Bmp) signalling. It is essential for primordial germ cell (PGC) development, for the development of the allantois and for amnion closure, as demonstrated by loss of Bmp signalling. By contrast, the appearance of ectopic PGC-like cells and regionalized ectopic vasculogenesis and haematopoiesis in thickened Smad5(m1/m1) amnion are amnion defects that have not been associated with loss of Bmp signalling components. We show that defects in amnion and allantois can already be detected at embryonic day (E) 7.5 in Smad5 mutant mice. However, ectopic Oct4-positive (Oct4(+)) and alkaline phosphatase-positive (AP(+)) cells appear suddenly in thickened amnion at E8.5, and at a remote distance from the allantois and posterior primitive streak, suggesting a change of fate in situ. These ectopic Oct4(+), AP(+) cells appear to be Stella negative and hence cannot be called bona fide PGCs. We demonstrate a robust upregulation of Bmp2 and Bmp4 expression, as well as of Erk and Smad activity, in the Smad5 mutant amnion. The ectopic expression of several Bmp target genes in different domains and the regionalized presence of cells of several Bmp-sensitive lineages in the mutant amnion suggest that different levels of Bmp signalling may determine cell fate. Injection of rBMP4 in the exocoelom of wild-type embryos can induce thickening of amnion, mimicking the early amnion phenotype in Smad5 mutants. These results support a model in which loss of Smad5 results paradoxically in gain of Bmp function defects in the amnion.


Amnion/embryology , Bone Morphogenetic Proteins/metabolism , DNA-Binding Proteins/physiology , Smad5 Protein/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/metabolism , Alleles , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Chimera , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Smad5 Protein/genetics , Trans-Activators/genetics , Transforming Growth Factor beta/genetics
15.
Am J Hum Genet ; 73(4): 926-32, 2003 Oct.
Article En | MEDLINE | ID: mdl-14508709

Slowed nerve-conduction velocities (NCVs) are a biological endophenotype in the majority of the hereditary motor and sensory neuropathies (HMSN). Here, we identified a family with autosomal dominant segregation of slowed NCVs without the clinical phenotype of HMSN. Peripheral-nerve biopsy showed predominantly thinly myelinated axons. We identified a locus at 8p23 and a Thr109Ile mutation in ARHGEF10, encoding a guanine-nucleotide exchange factor (GEF) for the Rho family of GTPase proteins (RhoGTPases). Rho GEFs are implicated in neural morphogenesis and connectivity and regulate the activity of small RhoGTPases by catalyzing the exchange of bound GDP by GTP. Expression analysis of ARHGEF10, by use of its mouse orthologue Gef10, showed that it is highly expressed in the peripheral nervous system. Our data support a role for ARHGEF10 in developmental myelination of peripheral nerves.


Chromosomes, Human, Pair 8 , Guanine Nucleotide Exchange Factors/genetics , Mutation , Myelin Sheath/physiology , Neural Conduction/genetics , Peripheral Nerves/physiology , Animals , Base Sequence , Chromosome Mapping , Female , Genetic Markers , Humans , Male , Mice , Molecular Sequence Data , Myelin Sheath/ultrastructure , Pedigree , RNA, Messenger/genetics , Rho Guanine Nucleotide Exchange Factors , Transcription, Genetic
16.
Genesis ; 37(1): 5-11, 2003 Sep.
Article En | MEDLINE | ID: mdl-14502571

Smad5 is a member of the Smad family of intracellular mediators of BMP signals and in endothelial cells of TGF-beta signals. We and others previously showed that loss of Smad5 in the mouse results in embryonic lethality (between E9.5-E11.5) due to multiple embryonic and extraembryonic defects. To circumvent the early embryonic lethality and to allow tissue- and time-specific Smad5 inactivation, we created a conditional Smad5 allele in the mouse. Floxed Smad5 (Smad5(flE2,Neo/flE2,Neo)) mice were generated in which both exon2 and the Neo-cassette were flanked by loxP sites. Here we demonstrate that embryos with ubiquitous Cre-mediated deletion of Smad5 (Smad5(flDeltaE2/flDeltaE2)) phenocopy the conventional Smad5 knockout mice. Smad5(flE2/flE2) mice are now available and will be a valuable tool to analyze the role of Smad5 beyond its crucial early embryonic function throughout development and postnatal life.


DNA-Binding Proteins/genetics , Mice, Knockout , Natriuretic Agents/metabolism , Phosphoproteins/genetics , Trans-Activators/genetics , Alleles , Animals , Binding Sites , Blotting, Southern , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Exons , Genetic Vectors , Heterozygote , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Polymerase Chain Reaction , Smad5 Protein , Stem Cells/cytology , Time Factors , Transforming Growth Factor beta/metabolism
17.
Am J Hum Genet ; 72(2): 465-70, 2003 Feb.
Article En | MEDLINE | ID: mdl-12522767

Recently, mutations in ZFHX1B, the gene that encodes Smad-interacting protein-1 (SIP1), were found to be implicated in the etiology of a dominant form of Hirschsprung disease-mental retardation syndrome in humans. To clarify the molecular mechanisms underlying the clinical features of SIP1 deficiency, we generated mice that bear a mutation comparable to those found in several human patients. Here, we show that Zfhx1b-knockout mice do not develop postotic vagal neural crest cells, the precursors of the enteric nervous system that is affected in patients with Hirschsprung disease, and they display a delamination arrest of cranial neural crest cells, which form the skeletomuscular elements of the vertebrate head. This suggests that Sip1 is essential for the development of vagal neural crest precursors and the migratory behavior of cranial neural crest in the mouse. Furthermore, we show that Sip1 is involved in the specification of neuroepithelium.


Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Intellectual Disability/genetics , Neural Crest/abnormalities , Repressor Proteins/genetics , Animals , Gene Expression , Homozygote , Mice , Mice, Knockout , Models, Genetic , Mutation , RNA, Messenger/metabolism , Zinc Finger E-box Binding Homeobox 2
...