Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Neurobiol Dis ; 199: 106564, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38876323

ABSTRACT

Biallelic variants in the SPG11 gene account for the most common form of autosomal recessive hereditary spastic paraplegia characterized by motor and cognitive impairment, with currently no therapeutic option. We previously observed in a Spg11 knockout mouse that neurodegeneration is associated with accumulation of gangliosides in lysosomes. To test whether a substrate reduction therapy could be a therapeutic option, we downregulated the key enzyme involved in ganglioside biosynthesis using an AAV-PHP.eB viral vector expressing a miRNA targeting St3gal5. Downregulation of St3gal5 in Spg11 knockout mice prevented the accumulation of gangliosides, delayed the onset of motor and cognitive symptoms, and prevented the upregulation of serum levels of neurofilament light chain, a biomarker widely used in neurodegenerative diseases. Importantly, similar results were observed when Spg11 knockout mice were administrated venglustat, a pharmacological inhibitor of glucosylceramide synthase expected to decrease ganglioside synthesis. Downregulation of St3gal5 or venglustat administration in Spg11 knockout mice strongly decreased the formation of axonal spheroids, previously associated with impaired trafficking. Venglustat had similar effect on cultured human SPG11 neurons. In conclusion, this work identifies the first disease-modifying therapeutic strategy in SPG11, and provides data supporting its relevance for therapeutic testing in SPG11 patients.

2.
Dev Dyn ; 253(1): 157-172, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37083132

ABSTRACT

BACKGROUND: Essential patterning processes transform the heart tube into a compartmentalized organ with distinct chambers separated by an atrioventricular canal (AVC). This transition involves the refinement of expression of genes that are first found broadly throughout the heart tube and then become restricted to the AVC. Despite the importance of cardiac patterning, we do not fully understand the mechanisms that limit gene expression to the AVC. RESULTS: We show that the zebrafish gene smarcc1a, encoding a BAF chromatin remodeling complex subunit homologous to mammalian BAF155, is critical for cardiac patterning. In smarcc1a mutants, myocardial differentiation and heart tube assembly appear to proceed normally. Subsequently, the smarcc1a mutant heart fails to exhibit refinement of gene expression patterns to the AVC, and the persistence of broad gene expression is accompanied by failure of chamber expansion. In addition to their cardiac defects, smarcc1a mutants lack pectoral fins, indicating similarity to tbx5a mutants. However, comparison of smarcc1a and tbx5a mutants suggests that perturbation of tbx5a function is not sufficient to cause the smarcc1a mutant phenotype. CONCLUSIONS: Our data indicate an important role for Smarcc1a-containing chromatin remodeling complexes in regulating the changes in gene expression and morphology that distinguish the AVC from the cardiac chambers.


Subject(s)
Endocardial Cushions , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Chromatin Assembly and Disassembly , Zebrafish Proteins/metabolism , Heart , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Mammals/metabolism
3.
PLoS Genet ; 19(10): e1010952, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37782669

ABSTRACT

Heterozygous de novo loss-of-function mutations in the gene expression regulator HNRNPU cause an early-onset developmental and epileptic encephalopathy. To gain insight into pathological mechanisms and lay the potential groundwork for developing targeted therapies, we characterized the neurophysiologic and cell-type-specific transcriptomic consequences of a mouse model of HNRNPU haploinsufficiency. Heterozygous mutants demonstrated global developmental delay, impaired ultrasonic vocalizations, cognitive dysfunction and increased seizure susceptibility, thus modeling aspects of the human disease. Single-cell RNA-sequencing of hippocampal and neocortical cells revealed widespread, yet modest, dysregulation of gene expression across mutant neuronal subtypes. We observed an increased burden of differentially-expressed genes in mutant excitatory neurons of the subiculum-a region of the hippocampus implicated in temporal lobe epilepsy. Evaluation of transcriptomic signature reversal as a therapeutic strategy highlights the potential importance of generating cell-type-specific signatures. Overall, this work provides insight into HNRNPU-mediated disease mechanisms and provides a framework for using single-cell RNA-sequencing to study transcriptional regulators implicated in disease.


Subject(s)
Haploinsufficiency , Transcriptome , Animals , Humans , Mice , Haploinsufficiency/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Neurons/metabolism , RNA/metabolism , Seizures/genetics , Transcriptome/genetics
4.
Front Cell Neurosci ; 17: 1175895, 2023.
Article in English | MEDLINE | ID: mdl-37275776

ABSTRACT

De novo mutations in GNB1, encoding the Gß1 subunit of G proteins, cause a neurodevelopmental disorder with global developmental delay and epilepsy, GNB1 encephalopathy. Here, we show that mice carrying a pathogenic mutation, K78R, recapitulate aspects of the disorder, including developmental delay and generalized seizures. Cultured mutant cortical neurons also display aberrant bursting activity on multi-electrode arrays. Strikingly, the antiepileptic drug ethosuximide (ETX) restores normal neuronal network behavior in vitro and suppresses spike-and-wave discharges (SWD) in vivo. ETX is a known blocker of T-type voltage-gated Ca2+ channels and G protein-coupled potassium (GIRK) channels. Accordingly, we present evidence that K78R results in a gain-of-function (GoF) effect by increasing the activation of GIRK channels in cultured neurons and a heterologous model (Xenopus oocytes)-an effect we show can be potently inhibited by ETX. This work implicates a GoF mechanism for GIRK channels in epilepsy, identifies a new mechanism of action for ETX in preventing seizures, and establishes this mouse model as a pre-clinical tool for translational research with predicative value for GNB1 encephalopathy.

5.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-34878101

ABSTRACT

The canonical Wnt/ß-catenin pathway governs a multitude of developmental processes in various cell lineages, including the melanocyte lineage. Indeed, ß-catenin regulates transcription of Mitf-M, the master regulator of this lineage. The first wave of melanocytes to colonize the skin is directly derived from neural crest cells, whereas the second wave of melanocytes is derived from Schwann cell precursors (SCPs). We investigated the influence of ß-catenin in the development of melanocytes of the first and second waves by generating mice expressing a constitutively active form of ß-catenin in cells expressing tyrosinase. Constitutive activation of ß-catenin did not affect the development of truncal melanoblasts but led to marked hyperpigmentation of the paws. By activating ß-catenin at various stages of development (E8.5-E11.5), we showed that the activation of ß-catenin in bipotent SCPs favored melanoblast specification at the expense of Schwann cells in the limbs within a specific temporal window. Furthermore, in vitro hyperactivation of the Wnt/ß-catenin pathway, which is required for melanocyte development, induces activation of Mitf-M, in turn repressing FoxD3 expression. In conclusion, ß-catenin overexpression promotes SCP cell fate decisions towards the melanocyte lineage.


Subject(s)
Cell Differentiation , Melanocytes/metabolism , Schwann Cells/cytology , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Lineage , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Melanocytes/cytology , Mice , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Protein Stability , Repressor Proteins/genetics , Repressor Proteins/metabolism , Schwann Cells/metabolism , Wnt Signaling Pathway , beta Catenin/genetics
6.
iScience ; 24(9): 103018, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34522861

ABSTRACT

Mutations in the GNB1 gene, encoding the Gß1 subunit of heterotrimeric G proteins, cause GNB1 Encephalopathy. Patients experience seizures, pointing to abnormal activity of ion channels or neurotransmitter receptors. We studied three Gß1 mutations (K78R, I80N and I80T) using computational and functional approaches. In heterologous expression models, these mutations did not alter the coupling between G protein-coupled receptors to Gi/o, or the Gßγ regulation of the neuronal voltage-gated Ca2+ channel CaV2.2. However, the mutations profoundly affected the Gßγ regulation of the G protein-gated inwardly rectifying potassium channels (GIRK, or Kir3). Changes were observed in Gß1 protein expression levels, Gßγ binding to cytosolic segments of GIRK subunits, and in Gßγ function, and included gain-of-function for K78R or loss-of-function for I80T/N, which were GIRK subunit-specific. Our findings offer new insights into subunit-dependent gating of GIRKs by Gßγ, and indicate diverse etiology of GNB1 Encephalopathy cases, bearing a potential for personalized treatment.

7.
Cell Rep ; 33(4): 108303, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33113364

ABSTRACT

Gain-of-function (GOF) variants in K+ channels cause severe childhood epilepsies, but there are no mechanisms to explain how increased K+ currents lead to network hyperexcitability. Here, we introduce a human Na+-activated K+ (KNa) channel variant (KCNT1-Y796H) into mice and, using a multiplatform approach, find motor cortex hyperexcitability and early-onset seizures, phenotypes strikingly similar to those of human patients. Although the variant increases KNa currents in cortical excitatory and inhibitory neurons, there is an increase in the KNa current across subthreshold voltages only in inhibitory neurons, particularly in those with non-fast-spiking properties, resulting in inhibitory-neuron-specific impairments in excitability and action potential (AP) generation. We further observe evidence of synaptic rewiring, including increases in homotypic synaptic connectivity, accompanied by network hyperexcitability and hypersynchronicity. These findings support inhibitory-neuron-specific mechanisms in mediating the epileptogenic effects of KCNT1 channel GOF, offering cell-type-specific currents and effects as promising targets for therapeutic intervention.


Subject(s)
Action Potentials/genetics , Epilepsy/genetics , GABAergic Neurons/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Sodium-Activated/metabolism , Seizures/genetics , Animals , Disease Models, Animal , Humans , Mice
8.
Am J Med Genet A ; 176(11): 2259-2275, 2018 11.
Article in English | MEDLINE | ID: mdl-30194818

ABSTRACT

De novo germline mutations in GNB1 have been associated with a neurodevelopmental phenotype. To date, 28 patients with variants classified as pathogenic have been reported. We add 18 patients with de novo mutations to this cohort, including a patient with mosaicism for a GNB1 mutation who presented with a milder phenotype. Consistent with previous reports, developmental delay in these patients was moderate to severe, and more than half of the patients were non-ambulatory and nonverbal. The most observed substitution affects the p.Ile80 residue encoded in exon 6, with 28% of patients carrying a variant at this residue. Dystonia and growth delay were observed more frequently in patients carrying variants in this residue, suggesting a potential genotype-phenotype correlation. In the new cohort of 18 patients, 50% of males had genitourinary anomalies and 61% of patients had gastrointestinal anomalies, suggesting a possible association of these findings with variants in GNB1. In addition, cutaneous mastocytosis, reported once before in a patient with a GNB1 variant, was observed in three additional patients, providing further evidence for an association to GNB1. We will review clinical and molecular data of these new cases and all previously reported cases to further define the phenotype and establish possible genotype-phenotype correlations.


Subject(s)
GTP-Binding Protein beta Subunits/genetics , Genetic Association Studies , Mutation/genetics , Adolescent , Child , Child, Preschool , Cohort Studies , Epilepsy/genetics , Female , GTP-Binding Protein beta Subunits/chemistry , Humans , Male , Nervous System/growth & development , Phenotype , Pregnancy , Protein Structure, Tertiary
9.
Development ; 145(3)2018 02 05.
Article in English | MEDLINE | ID: mdl-29361575

ABSTRACT

NKX2-5 is the most commonly mutated gene associated with human congenital heart defects (CHDs), with a predilection for cardiac pole abnormalities. This homeodomain transcription factor is a central regulator of cardiac development and is expressed in both the first and second heart fields (FHF and SHF). We have previously revealed essential functions of nkx2.5 and nkx2.7, two Nkx2-5 homologs expressed in zebrafish cardiomyocytes, in maintaining ventricular identity. However, the differential roles of these genes in the specific subpopulations of the anterior (aSHF) and posterior (pSHF) SHFs have yet to be fully defined. Here, we show that Nkx genes regulate aSHF and pSHF progenitors through independent mechanisms. We demonstrate that Nkx genes restrict proliferation of aSHF progenitors in the outflow tract, delimit the number of pSHF progenitors at the venous pole and pattern the sinoatrial node acting through Isl1 repression. Moreover, optical mapping highlights the requirement for Nkx gene dose in establishing electrophysiological chamber identity and in integrating the physiological connectivity of FHF and SHF cardiomyocytes. Ultimately, our results may shed light on the discrete errors responsible for NKX2-5-dependent human CHDs of the cardiac outflow and inflow tracts.


Subject(s)
Heart/embryology , Homeobox Protein Nkx-2.5/genetics , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Developmental , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Humans , Mutation
10.
Genesis ; 55(3)2017 03.
Article in English | MEDLINE | ID: mdl-28109039

ABSTRACT

In gnathostomes, dorsoventral (D-V) patterning of neural crest cells (NCC) within the pharyngeal arches is crucial for the development of hinged jaws. One of the key signals that mediate this process is Endothelin-1 (EDN1). Loss of EDN1 binding to the Endothelin-A receptor (EDNRA) results in loss of EDNRA signaling and subsequent facial birth defects in humans, mice and zebrafish. A rate-limiting step in this crucial signaling pathway is the conversion of immature EDN1 into a mature active form by Endothelin converting enzyme-1 (ECE1). However, surprisingly little is known about how Ece1 transcription is induced or regulated. We show here that Nkx2.5 is required for proper craniofacial development in zebrafish and acts in part by upregulating ece1 expression. Disruption of nkx2.5 in zebrafish embryos results in defects in both ventral and dorsal pharyngeal arch-derived elements, with changes in ventral arch gene expression consistent with a disruption in Ednra signaling. ece1 mRNA rescues the nkx2.5 morphant phenotype, indicating that Nkx2.5 functions through modulating Ece1 expression or function. These studies illustrate a new function for Nkx2.5 in embryonic development and provide new avenues with which to pursue potential mechanisms underlying human facial disorders.


Subject(s)
Endothelin-Converting Enzymes/genetics , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5/genetics , Neural Crest/metabolism , Zebrafish Proteins/genetics , Animals , Endothelin-Converting Enzymes/metabolism , Homeobox Protein Nkx-2.5/metabolism , Mice , Neural Crest/embryology , Pharynx/embryology , Pharynx/metabolism , Up-Regulation , Zebrafish , Zebrafish Proteins/metabolism
11.
Am J Hum Genet ; 98(5): 1001-1010, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27108799

ABSTRACT

Whole-exome sequencing of 13 individuals with developmental delay commonly accompanied by abnormal muscle tone and seizures identified de novo missense mutations enriched within a sub-region of GNB1, a gene encoding the guanine nucleotide-binding protein subunit beta-1, Gß. These 13 individuals were identified among a base of 5,855 individuals recruited for various undiagnosed genetic disorders. The probability of observing 13 or more de novo mutations by chance among 5,855 individuals is very low (p = 7.1 × 10(-21)), implicating GNB1 as a genome-wide-significant disease-associated gene. The majority of these 13 mutations affect known Gß binding sites, which suggests that a likely disease mechanism is through the disruption of the protein interface required for Gα-Gßγ interaction (resulting in a constitutively active Gßγ) or through the disruption of residues relevant for interaction between Gßγ and certain downstream effectors (resulting in reduced interaction with the effectors). Strikingly, 8 of the 13 individuals recruited here for a neurodevelopmental disorder have a germline de novo GNB1 mutation that overlaps a set of five recurrent somatic tumor mutations for which recent functional studies demonstrated a gain-of-function effect due to constitutive activation of G protein downstream signaling cascades for some of the affected residues.


Subject(s)
Developmental Disabilities/etiology , GTP-Binding Protein beta Subunits/genetics , Germ-Line Mutation/genetics , Intellectual Disability/etiology , Muscle Hypotonia/etiology , Seizures/etiology , Adolescent , Adult , Child , Child, Preschool , Developmental Disabilities/pathology , Exome/genetics , Female , GTP-Binding Protein beta Subunits/chemistry , Humans , Infant , Intellectual Disability/pathology , Male , Muscle Hypotonia/pathology , Phenotype , Protein Conformation , Seizures/pathology , Signal Transduction , Young Adult
12.
Nat Commun ; 6: 8093, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26307673

ABSTRACT

Loss of the tumour suppressor PTEN is frequent in human melanoma, results in MAPK activation, suppresses senescence and mediates metastatic behaviour. How PTEN loss mediates these effects is unknown. Here we show that loss of PTEN in epithelial and melanocytic cell lines induces the nuclear localization and transcriptional activation of ß-catenin independent of the PI3K-AKT-GSK3ß axis. The absence of PTEN leads to caveolin-1 (CAV1)-dependent ß-catenin transcriptional modulation in vitro, cooperates with NRAS(Q61K) to initiate melanomagenesis in vivo and induces efficient metastasis formation associated with E-cadherin internalization. The CAV1-ß-catenin axis is mediated by a feedback loop in which ß-catenin represses transcription of miR-199a-5p and miR-203, which suppress the levels of CAV1 mRNA in melanoma cells. These data reveal a mechanism by which loss of PTEN increases CAV1-mediated dissociation of ß-catenin from membranous E-cadherin, which may promote senescence bypass and metastasis.


Subject(s)
Cadherins/metabolism , Caveolin 1/genetics , Melanocytes/metabolism , Melanoma/genetics , PTEN Phosphohydrolase/genetics , Skin Neoplasms/genetics , Transcriptional Activation/genetics , beta Catenin/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Feedback, Physiological , GTP Phosphohydrolases/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , Melanoma/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , MicroRNAs , Microscopy, Fluorescence , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/metabolism
13.
Dev Biol ; 400(1): 10-22, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25536398

ABSTRACT

Temporally controlled mechanisms that define the unique features of ventricular and atrial cardiomyocyte identities are essential for the construction of a coordinated, morphologically intact heart. We have previously demonstrated an important role for nkx genes in maintaining ventricular identity, however, the specific timing of nkx2.5 function in distinct cardiomyocyte populations has yet to be elucidated. Here, we show that heat-shock induction of a novel transgenic line, Tg(hsp70l:nkx2.5-EGFP), during the initial stages of cardiomyocyte differentiation leads to rescue of chamber shape and identity in nkx2.5(-/-) embryos as chambers emerge. Intriguingly, our findings link an early role of this essential cardiac transcription factor with a later function. Moreover, these data reveal that nkx2.5 is also required in the second heart field as the heart tube forms, reflecting the temporal delay in differentiation of this population. Thus, our results support a model in which nkx genes induce downstream targets that are necessary to maintain chamber-specific identity in both early- and late-differentiating cardiomyocytes at discrete stages in cardiac morphogenesis. Furthermore, we show that overexpression of nkx2.5 during the first and second heart field development not only rescues the mutant phenotype, but also is sufficient for proper function of the adult heart. Taken together, these results shed new light on the stage-dependent mechanisms that sculpt chamber-specific cardiomyocytes and, therefore, have the potential to improve in vitro generation of ventricular cells to treat myocardial infarction and congenital heart disease.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Heart Ventricles/embryology , Morphogenesis/physiology , Myocytes, Cardiac/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Blotting, Western , Cell Count , Cell Differentiation/physiology , DNA Primers/genetics , Gene Knockout Techniques , Genotype , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.5 , Image Processing, Computer-Assisted , In Situ Hybridization , Microscopy, Fluorescence , Polymerase Chain Reaction , Transcription Factors/genetics , Zebrafish Proteins/genetics
14.
Development ; 140(20): 4203-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24026123

ABSTRACT

Establishment of specific characteristics of each embryonic cardiac chamber is crucial for development of a fully functional adult heart. Despite the importance of defining and maintaining unique features in ventricular and atrial cardiomyocytes, the regulatory mechanisms guiding these processes are poorly understood. Here, we show that the homeodomain transcription factors Nkx2.5 and Nkx2.7 are necessary to sustain ventricular chamber attributes through repression of atrial chamber identity. Mutation of nkx2.5 in zebrafish yields embryos with diminutive ventricular and bulbous atrial chambers. These chamber deformities emerge gradually during development, with a severe collapse in the number of ventricular cardiomyocytes and an accumulation of excess atrial cardiomyocytes as the heart matures. Removal of nkx2.7 function from nkx2.5 mutants exacerbates the loss of ventricular cells and the gain of atrial cells. Moreover, in these Nkx-deficient embryos, expression of vmhc, a ventricular gene, fades, whereas expression of amhc, an atrial gene, expands. Cell-labeling experiments suggest that ventricular cardiomyocytes can transform into atrial cardiomyocytes in the absence of Nkx gene function. Through suggestion of transdifferentiation from ventricular to atrial fate, our data reveal a pivotal role for Nkx genes in maintaining ventricular identity and highlight remarkable plasticity in differentiated myocardium. Thus, our results are relevant to the etiologies of fetal and neonatal cardiac pathology and could direct future innovations in cardiac regenerative medicine.


Subject(s)
Heart Atria/embryology , Heart Ventricles/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Atrial Myosins/biosynthesis , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Developmental , Genotype , Heart Atria/metabolism , Heart Ventricles/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mutation , Myocytes, Cardiac/metabolism , Transcription Factors/genetics , Transcription, Genetic , Ventricular Myosins/biosynthesis , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
15.
PLoS One ; 8(1): e53183, 2013.
Article in English | MEDLINE | ID: mdl-23382837

ABSTRACT

BACKGROUND: Patent ductus arteriosus is a life-threatening condition frequent in premature newborns but also present in some term infants. Current mouse models of this malformation generally lead to perinatal death, not reproducing the full phenotypic spectrum in humans, in whom genetic inheritance appears complex. The ductus arteriosus (DA), a temporary fetal vessel that bypasses the lungs by shunting the aortic arch to the pulmonary artery, is constituted by smooth muscle cells of distinct origins (SMC1 and SMC2) and many fewer melanocytes. To understand novel mechanisms preventing DA closure at birth, we evaluated the importance of cell fate specification in SMC that form the DA during embryonic development. Upon specific Tyr::Cre-driven activation of Wnt/ß-catenin signaling at the time of cell fate specification, melanocytes replaced the SMC2 population of the DA, suggesting that SMC2 and melanocytes have a common precursor. The number of SMC1 in the DA remained similar to that in controls, but insufficient to allow full DA closure at birth. Thus, there was no cellular compensation by SMC1 for the loss of SMC2. Mice in which only melanocytes were genetically ablated after specification from their potential common precursor with SMC2, demonstrated that differentiated melanocytes themselves do not affect DA closure. Loss of the SMC2 population, independent of the presence of melanocytes, is therefore a cause of patent ductus arteriosus and premature death in the first months of life. Our results indicate that patent ductus arteriosus can result from the insufficient differentiation, proliferation, or contractility of a specific smooth muscle subpopulation that shares a common neural crest precursor with cardiovascular melanocytes.


Subject(s)
Cell Differentiation/physiology , Ductus Arteriosus, Patent/physiopathology , Embryonic Development , Myocytes, Smooth Muscle/pathology , Premature Birth/physiopathology , Animals , Cell Lineage , Cell Proliferation , Ductus Arteriosus, Patent/etiology , Female , Humans , Melanocytes/cytology , Mice , Muscle Contraction/physiology , Pregnancy , Wnt Signaling Pathway
16.
Mol Cell Biol ; 32(7): 1237-47, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22290434

ABSTRACT

MITF-M and PAX3 are proteins central to the establishment and transformation of the melanocyte lineage. They control various cellular mechanisms, including migration and proliferation. BRN2 is a POU domain transcription factor expressed in melanoma cell lines and is involved in proliferation and invasion, at least in part by regulating the expression of MITF-M and PAX3. The T361 and S362 residues of BRN2, both in the POU domain, are conserved throughout the POU protein family and are targets for phosphorylation, but their roles in vivo remain unknown. To examine the role of this phosphorylation, we generated mutant BRN2 in which these two residues were replaced with alanines (BRN2TS→BRN2AA). When expressed in melanocytes in vitro or in the melanocyte lineage in transgenic mice, BRN2TS induced proliferation and repressed migration, whereas BRN2AA repressed both proliferation and migration. BRN2TS and BRN2AA bound and repressed the MITF-M promoter, whereas PAX3 transcription was induced by BRN2TS but repressed by BRN2AA. Expression of the BRN2AA transgene in a Mitf heterozygous background and in a Pax3 mutant background enhanced the coat color phenotype. Our findings show that melanocyte migration and proliferation are controlled both through the regulation of PAX3 by nonphosphorylated BRN2 and through the regulation of MITF-M by the overall BRN2 level.


Subject(s)
Cell Proliferation , Melanocytes/cytology , Nerve Tissue Proteins/metabolism , POU Domain Factors/metabolism , Paired Box Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Movement , Humans , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Mutation , Nerve Tissue Proteins/genetics , PAX3 Transcription Factor , POU Domain Factors/genetics , Phenotype , Phosphorylation , Promoter Regions, Genetic , Transcription, Genetic
18.
J Invest Dermatol ; 132(1): 170-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21850021

ABSTRACT

Studying the development of melanoblasts, precursors of melanocytes, is challenging owing to their scarcity and dispersion in the skin embryo. However, this is an important subject because diverse diseases are associated with defective melanoblast development. Consequently, characterizing patterns of expression in melanoblasts during normal development is an important issue. This requires isolating enough melanoblasts during embryonic development to obtain sufficient RNA to study their transcriptome. ZEG reporter mouse line crossed with Tyr::Cre mouse line was used to label melanoblasts by enhanced green fluorescent protein (EGFP) autofluorescence. We isolated melanoblasts by FACS from the skin of E14.5-E16.5 embryos, and obtained sufficient cells for large-scale transcriptomic analysis after RNA isolation and amplification. We confirmed our array-based data for various genes of interest by standard quantitative real-time RT-PCR. We demonstrated that phosphatase and tensin homolog was expressed in melanoblasts but BRN2 was not, although both are involved in melanomagenesis. We also revealed the potential contribution of genes not previously implicated in any function in melanocytes or even in neural crest derivatives. Finally, the Schwann cell markers, PLP1 and FABP7, were significantly expressed in melanoblasts, melanocytes, and melanoma. This study demonstrates the feasibility of the transcriptomic analysis of purified melanoblasts at different embryonic stages and reveals the involvement of previously unreported genes in melanoblast development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/physiology , Melanocytes/cytology , Skin/embryology , Transcriptome/physiology , Animals , Cell Line , Cell Separation/methods , Fetus/cytology , Fetus/physiology , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , POU Domain Factors/genetics , PTEN Phosphohydrolase/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/methods , Skin/cytology
19.
Genesis ; 48(2): 96-100, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20014334

ABSTRACT

The Z/EG transgenic mouse line, produced by Novak et al., displays tissue-specific EGFP expression after Cre-mediated recombination. The autofluorescence of EGFP allows the visualization of cells of interest displaying Cre recombination. The initial construct was designed such that cells without Cre recombination express the beta-galactosidase marker, facilitating counterselection. We used inverse PCR to identify the site of integration of the Z/EG transgene, to improve the efficiency of homozygous Z/EG mouse production. Recombined cells produced large amounts of EGFP protein, resulting in higher levels of fluorescence and therefore greater contrast with nonrecombined cells. We mapped the transgene to the G1 region of chromosome 5. This random insertion was found to have occurred 230-bp upstream from the start codon of the Rasa4 gene. The insertion of the Z/EG transgene in the C57BL/6 genetic background had no effect on Rasa4 expression. Homozygous Z/EG mice therefore had no obvious phenotype.


Subject(s)
Genome , Transgenes/genetics , Animals , Cell Line , Chromosome Mapping , Chromosomes, Mammalian , Fluorescent Dyes/metabolism , Gene Expression , Genetic Markers , Green Fluorescent Proteins/metabolism , Homozygote , Mice , Mice, Inbred C57BL , Mice, Transgenic , beta-Galactosidase/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...