Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 292(31): 13045-13055, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28623229

ABSTRACT

Transcription factors play crucial roles in patterning posterior neuroectoderm. Previously, zinc finger transcription factor znfl1 was reported to be expressed in the posterior neuroectoderm of zebrafish embryos. However, its roles remain unknown. Here, we report that there are 13 copies of znfl1 in the zebrafish genome, and all the paralogues share highly identical protein sequences and cDNA sequences. When znfl1s are knocked down using a morpholino to inhibit their translation or dCas9-Eve to inhibit their transcription, the zebrafish gastrula displays reduced expression of hoxb1b, the marker gene for the posterior neuroectoderm. Further analyses reveal that diminishing znfl1s produces the decreased expressions of pou5f3, whereas overexpression of pou5f3 effectively rescues the reduced expression of hoxb1b in the posterior neuroectoderm. Additionally, knocking down znfl1s causes the reduced expression of sall4, a direct regulator of pou5f3, in the posterior neuroectoderm, and overexpression of sall4 rescues the expression of pou5f3 in the knockdown embryos. In contrast, knocking down either pou5f3 or sall4 does not affect the expressions of znfl1s Taken together, our results demonstrate that zebrafish znfl1s control the expression of hoxb1b in the posterior neuroectoderm by acting upstream of pou5f3 and sall4.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Plate/metabolism , Octamer Transcription Factor-3/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Animals , Biomarkers/metabolism , Computational Biology , Gastrula/drug effects , Gastrula/metabolism , Gene Dosage , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , In Situ Hybridization , Microinjections , Morpholinos/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neural Plate/drug effects , Neural Plate/embryology , Neurogenesis/drug effects , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , RNA Interference , RNA, Antisense/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
2.
Stem Cell Reports ; 6(5): 679-691, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27132889

ABSTRACT

Human pluripotent stem cells (hPSCs) exist in heterogeneous micro-environments with multiple subpopulations, convoluting fate-regulation analysis. We patterned hPSCs into engineered micro-environments and screened responses to 400 small-molecule kinase inhibitors, measuring yield and purity outputs of undifferentiated, neuroectoderm, mesendoderm, and extra-embryonic populations. Enrichment analysis revealed mammalian target of rapamycin (mTOR) inhibition as a strong inducer of mesendoderm. Dose responses of mTOR inhibitors such as rapamycin synergized with Bone Morphogenetic protein 4 (BMP4) and activin A to enhance the yield and purity of BRACHYURY-expressing cells. Mechanistically, small interfering RNA knockdown of RAPTOR, a component of mTOR complex 1, phenocopied the mesendoderm-enhancing effects of rapamycin. Functional analysis during mesoderm and endoderm differentiation revealed that mTOR inhibition increased the output of hemogenic endothelial cells 3-fold, with a concomitant enhancement of blood colony-forming cells. These data demonstrate the power of our multi-lineage screening approach and identify mTOR signaling as a node in hPSC differentiation to mesendoderm and its derivatives.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Pluripotent Stem Cells/drug effects , Activins/genetics , Bone Morphogenetic Protein 4/genetics , Cellular Microenvironment/drug effects , Endoderm/drug effects , Endoderm/metabolism , Enzyme Inhibitors/pharmacology , Fetal Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Neural Plate/cytology , Neural Plate/drug effects , Neural Plate/metabolism , Phosphotransferases/antagonists & inhibitors , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Regulatory-Associated Protein of mTOR/genetics , Sirolimus/pharmacology , Small Molecule Libraries/pharmacology , T-Box Domain Proteins/genetics
3.
Dis Model Mech ; 9(6): 607-20, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27101845

ABSTRACT

Of all live births with congenital anomalies, approximately one-third exhibit deformities of the head and face. Most craniofacial disorders are associated with defects in a migratory stem and progenitor cell population, which is designated the neural crest (NC). Musculocontractural Ehlers-Danlos syndrome (MCEDS) is a heritable connective tissue disorder with distinct craniofacial features; this syndrome comprises multiple congenital malformations that are caused by dysfunction of dermatan sulfate (DS) biosynthetic enzymes, including DS epimerase-1 (DS-epi1; also known as DSE). Studies in mice have extended our understanding of DS-epi1 in connective tissue maintenance; however, its role in fetal development is not understood. We demonstrate that DS-epi1 is important for the generation of isolated iduronic acid residues in chondroitin sulfate (CS)/DS proteoglycans in early Xenopus embryos. The knockdown of DS-epi1 does not affect the formation of early NC progenitors; however, it impairs the correct activation of transcription factors involved in the epithelial-mesenchymal transition (EMT) and reduces the extent of NC cell migration, which leads to a decrease in NC-derived craniofacial skeleton, melanocytes and dorsal fin structures. Transplantation experiments demonstrate a tissue-autonomous role for DS-epi1 in cranial NC cell migration in vivo Cranial NC explant and single-cell cultures indicate a requirement of DS-epi1 in cell adhesion, spreading and extension of polarized cell processes on fibronectin. Thus, our work indicates a functional link between DS and NC cell migration. We conclude that NC defects in the EMT and cell migration might account for the craniofacial anomalies and other congenital malformations in MCEDS, which might facilitate the diagnosis and development of therapies for this distressing condition. Moreover, the presented correlations between human DS-epi1 expression and gene sets of mesenchymal character, invasion and metastasis in neuroblastoma and malignant melanoma suggest an association between DS and NC-derived cancers.


Subject(s)
Cell Movement/drug effects , Dermatan Sulfate/pharmacology , Ehlers-Danlos Syndrome/pathology , Fibronectins/metabolism , Muscles/pathology , Neural Crest/pathology , Animals , Base Sequence , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Polarity , Chondroitin Sulfates/metabolism , Ehlers-Danlos Syndrome/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Feedback, Physiological , Gene Expression Regulation, Developmental , Iduronic Acid/metabolism , Models, Biological , Neoplasms/pathology , Neural Plate/drug effects , Neural Plate/metabolism , Racemases and Epimerases/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics
4.
In Vitro Cell Dev Biol Anim ; 52(1): 35-44, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26288008

ABSTRACT

We have investigated the role chondroitin sulfate has on cell interactions during neural plate formation in the early chick embryo. Using tissue culture isolates from the prospective neural plate, we have measured neural gene expression profiles associated with neural stem cell differentiation. Removal of chondroitin sulfate from stage 4 neural plate tissue leads to altered associations of N-cadherin-positive neural progenitors and causes changes in the normal sequence of neural marker gene expression. Absence of chondroitin sulfate in the neural plate leads to reduced Sox2 expression and is accompanied by an increase in the expression of anterior markers of neural regionalization. Results obtained in this study suggest that the presence of chondroitin sulfate in the anterior chick embryo is instrumental in maintaining cells in the neural precursor state.


Subject(s)
Cell Communication/drug effects , Cell Differentiation/genetics , Chondroitin Sulfates/pharmacology , Neural Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Neural Plate/drug effects , Neural Plate/growth & development , SOXB1 Transcription Factors/biosynthesis
5.
Arch Toxicol ; 89(9): 1599-618, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26272509

ABSTRACT

Test systems to identify developmental toxicants are urgently needed. A combination of human stem cell technology and transcriptome analysis was to provide a proof of concept that toxicants with a related mode of action can be identified and grouped for read-across. We chose a test system of developmental toxicity, related to the generation of neuroectoderm from pluripotent stem cells (UKN1), and exposed cells for 6 days to the histone deacetylase inhibitors (HDACi) valproic acid, trichostatin A, vorinostat, belinostat, panobinostat and entinostat. To provide insight into their toxic action, we identified HDACi consensus genes, assigned them to superordinate biological processes and mapped them to a human transcription factor network constructed from hundreds of transcriptome data sets. We also tested a heterogeneous group of 'mercurials' (methylmercury, thimerosal, mercury(II)chloride, mercury(II)bromide, 4-chloromercuribenzoic acid, phenylmercuric acid). Microarray data were compared at the highest non-cytotoxic concentration for all 12 toxicants. A support vector machine (SVM)-based classifier predicted all HDACi correctly. For validation, the classifier was applied to legacy data sets of HDACi, and for each exposure situation, the SVM predictions correlated with the developmental toxicity. Finally, optimization of the classifier based on 100 probe sets showed that eight genes (F2RL2, TFAP2B, EDNRA, FOXD3, SIX3, MT1E, ETS1 and LHX2) are sufficient to separate HDACi from mercurials. Our data demonstrate how human stem cells and transcriptome analysis can be combined for mechanistic grouping and prediction of toxicants. Extension of this concept to mechanisms beyond HDACi would allow prediction of human developmental toxicity hazard of unknown compounds with the UKN1 test system.


Subject(s)
Histone Deacetylase Inhibitors/toxicity , Neural Plate/drug effects , Pluripotent Stem Cells/drug effects , Transcriptome , Gene Expression Profiling , Humans , Neural Plate/metabolism , Oligonucleotide Array Sequence Analysis
6.
Environ Sci Technol ; 49(16): 10165-72, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26182351

ABSTRACT

Extraction of petrochemicals from the surface mining of oil sand deposits results in generation of large volumes of oil sands process-affected water (OSPW). Naphthenic acids (NA) are generally considered to be among the most toxic components of OSPW. Previous studies have shown that NAs are toxic to aquatic organisms, however knowledge of their effects on mammalian health and development is limited. In the present study, we evaluated the developmental effects of an NA extract prepared from fresh OSPW on differentiating mouse embryonic stem cells (ESC). We found that treatment of differentiating cells with the NA extract at noncytotoxic concentrations alters expression of various lineage specification markers and development of the heart. Notably, expression of cardiac specific markers such as Nkx2.5, Gata4, and Mef2c were significantly up-regulated. Moreover, exposure to the NA extract enhanced differentiation of embryoid bodies and resulted in the early appearance of spontaneously beating clusters. Interestingly, exposure of undifferentiated mouse ESCs to the NA extract did not change the expression level of pluripotency markers (i.e., Oct4, Nanog, and Sox2). Altogether, these data identify some of the molecular pathways affected by components within this NA extract during differentiation of mammalian cells.


Subject(s)
Carboxylic Acids/toxicity , Cell Differentiation/drug effects , Heart/embryology , Mouse Embryonic Stem Cells/cytology , Oil and Gas Fields , Water Pollutants, Chemical/toxicity , Animals , Biomarkers/metabolism , Cell Death/drug effects , Cell Lineage/drug effects , Heart/drug effects , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neural Plate/drug effects , Neural Plate/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
7.
Mol Brain ; 7: 67, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25223405

ABSTRACT

BACKGROUND: Alcohol is detrimental to early development. Fetal alcohol spectrum disorders (FASD) due to maternal alcohol abuse results in a series of developmental abnormalities including cranial facial dysmorphology, ocular anomalies, congenital heart defects, microcephaly and intellectual disabilities. Previous studies have been shown that ethanol exposure causes neural crest (NC) apoptosis and perturbation of neural crest migration. However, the underlying mechanism remains elusive. In this report we investigated the fetal effect of alcohol on the process of neural crest development in the Xenopus leavis. RESULTS: Pre-gastrulation exposure of 2-4% alcohol induces apoptosis in Xenopus embryo whereas 1% alcohol specifically impairs neural crest migration without observing discernible apoptosis. Additionally, 1% alcohol treatment considerably increased the phenotype of small head (43.4% ± 4.4%, total embryo n = 234), and 1.5% and 2.0% dramatically augment the deformation to 81.2% ± 6.5% (n = 205) and 91.6% ± 3.0% (n = 235), respectively (P < 0.05). Significant accumulation of Homocysteine was caused by alcohol treatment in embryos and 5-mehtyltetrahydrofolate restores neural crest migration and alleviates homocysteine accumulation, resulting in inhibition of the alcohol-induced neurocristopathies. CONCLUSIONS: Our study demonstrates that prenatal alcohol exposure causes neural crest cell migration abnormality and 5-mehtyltetrahydrofolate could be beneficial for treating FASD.


Subject(s)
Cell Movement/drug effects , Ethanol/toxicity , Neural Crest/pathology , Tetrahydrofolates/pharmacology , Animals , Apoptosis/drug effects , Cartilage/drug effects , Cartilage/embryology , Models, Animal , Neural Crest/drug effects , Neural Plate/drug effects , Neural Plate/pathology , Pigments, Biological/metabolism , Xenopus laevis/embryology
8.
Exp Cell Res ; 328(2): 361-78, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25193078

ABSTRACT

JMJD2A is a lysine trimethyl-specific histone demethylase that is highly expressed in a variety of tumours. The role of JMJD2A in tumour progression remains unclear. The objectives of this study were to identify JMJD2A-regulated genes and understand the function of JMJD2A in p53-null neuroectodermal stem cells (p53(-/-) NE-4Cs). We determined the effect of LPS as a model of inflammation in p53(-/-) NE-4Cs and investigated whether the epigenetic modifier JMJD2A alter the expression of tumourigenic inflammatory genes. Global gene expression was measured in JMJD2A knockdown (kd) p53(-/-) NE-4Cs and in LPS-stimulated JMJD2A-kd p53(-/-) NE-4C cells. JMJD2A attenuation significantly down-regulated genes were Cdca2, Ccnd2, Ccnd1, Crebbp, IL6rα, and Stat3 related with cell cycle, proliferation, and inflammatory-disease responses. Importantly, some tumour-suppressor genes including Dapk3, Timp2 and TFPI were significantly up-regulated but were not affected by silencing of the JMJD2B. Furthermore, we confirmed the attenuation of JMJD2A also down-regulated Cdca2, Ccnd2, Crebbp, and Rest in primary NSCs isolated from the forebrains of E15 embryos of C57/BL6J mice with effective p53 inhibitor pifithrin-α (PFT-α). Transcription factor (TF) motif analysis revealed known binding patterns for CDC5, MYC, and CREB, as well as three novel motifs in JMJD2A-regulated genes. IPA established molecular networks. The molecular network signatures and functional gene-expression profiling data from this study warrants further investigation as an effective therapeutic target, and studies to elucidate the molecular mechanism of JMJD2A-kd-dependent effects in neuroectodermal stem cells should be performed.


Subject(s)
Carcinogenesis/genetics , Cell Cycle/genetics , Histone Demethylases/genetics , Inflammation/genetics , Lipopolysaccharides/pharmacology , Neural Plate/metabolism , Stem Cells/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Down-Regulation/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Genes, Tumor Suppressor , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Neural Plate/drug effects , Stem Cells/drug effects , Transcription Factors/genetics , Transcriptome/genetics , Tumor Suppressor Protein p53/genetics
9.
Cell Commun Signal ; 12: 19, 2014 Mar 23.
Article in English | MEDLINE | ID: mdl-24656144

ABSTRACT

Prostaglandin E2 (PGE2) is a natural lipid-derived molecule that is involved in important physiological functions. Abnormal PGE2 signalling has been associated with pathologies of the nervous system. Previous studies provide evidence for the interaction of PGE2 and canonical Wnt signalling pathways in non-neuronal cells. Since the Wnt pathway is crucial in the development and organization of the brain, the main goal of this study is to determine whether collaboration between these pathways exists in neuronal cell types. We report that PGE2 interacts with canonical Wnt signalling through PKA and PI-3K in neuroectodermal (NE-4C) stem cells. We used time-lapse microscopy to determine that PGE2 increases the final distance from origin, path length travelled, and the average speed of migration in Wnt-activated cells. Furthermore, PGE2 alters distinct cellular phenotypes that are characteristic of Wnt-induced NE-4C cells, which corresponds to the modified splitting behaviour of the cells. We also found that in Wnt-induced cells the level of ß-catenin protein was increased and the expression levels of Wnt-target genes (Ctnnb1, Ptgs2, Ccnd1, Mmp9) was significantly upregulated in response to PGE2 treatment. This confirms that PGE2 activated the canonical Wnt signalling pathway. Furthermore, the upregulated genes have been previously associated with ASD. Our findings show, for the first time, evidence for cross-talk between PGE2 and Wnt signalling in neuronal cells, where PKA and PI-3K might act as mediators between the two pathways. Given the importance of PGE2 and Wnt signalling in prenatal development of the nervous system, our study provides insight into how interaction between these two pathways may influence neurodevelopment.


Subject(s)
Cell Movement , Cell Proliferation , Dinoprostone/pharmacology , Neural Plate/drug effects , Neural Stem Cells/drug effects , Wnt Signaling Pathway , Animals , Cell Line , Child Development Disorders, Pervasive/etiology , Child Development Disorders, Pervasive/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Neural Plate/cytology , Neural Plate/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Comp Med ; 64(2): 140-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24674590

ABSTRACT

Retinoic acid is a widely used factor in both mouse and human embryonic stem cells. It suppresses differentiation to mesoderm and enhances differentiation to ectoderm. Fibroblast growth factor 2 (FGF2) is widely used to induce differentiation to neurons in mice, yet in primates, including humans, it maintains embryonic stem cells in the undifferentiated state. In this study, we established an FGF2 low-dose-dependent embryonic stem cell line from cynomolgus monkeys and then analyzed neural differentiation in cultures supplemented with retinoic acid and FGF2. When only retinoic acid was added to culture, neurons differentiated from FGF2 low-dose-dependent embryonic stem cells. When both retinoic acid and FGF2 were added, neurons and astrocytes differentiated from the same embryonic stem cell line. Thus, retinoic acid promotes the differentiation from embryonic stem cells to neuroectoderm. Although FGF2 seems to promote self-renewal in stem cells, its effects on the differentiation of stem cells are influenced by the presence or absence of supplemental retinoic acid.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Macaca fascicularis/physiology , Neural Plate/drug effects , Tretinoin/pharmacology , Animals , Dose-Response Relationship, Drug , Embryonic Stem Cells/physiology , Karyotyping , Microscopy, Fluorescence , Neural Plate/physiology
11.
Curr Med Chem ; 21(8): 1017-25, 2014.
Article in English | MEDLINE | ID: mdl-23992320

ABSTRACT

Advanced enteropancreatic (EP) neuroendocrine tumors (NETs) can be treated with several different therapies, including chemotherapy, biotherapy, and locoregional treatments. Over the last few decades, impressive progress has been made in the biotherapy field. Three main druggable molecular targets have been studied and developed in terms of therapy: somatostatin receptor (sstr), mammalian target of rapamycin (mTOR), and angiogenic factors. In particular, research has moved from the old somatostatin analogs (SSAs), such as octreotide (OCT) and lanreotide (LAN), specifically binding to the sstr-2, to the newer pasireotide (PAS), which presents a wider sstr spectrum. Over the last ten years, several molecular targeted agents (MTAs) have been studied in phase II trials, and very few of them have reached phase III. The mTOR inhibitor everolimus and the multitargeted inhibitor sunitinib have been approved for clinical use by the FDA and EMA in advanced well/moderately-differentiated (WD, MD) progressive pancreatic neuroendocrine tumors (PNETs), on the basis of the positive results of two international large randomized phase III trials vs. placebo. Bevacizumab has been studied in a large US phase III trial vs. interferon (IFN)-alfa2b, and results are pending. In this review, the biological and clinical aspects of MTAs introduced into clinical practice or which are currently in an advanced phase of clinical investigation are addressed.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy/methods , Neural Plate/pathology , Neuroendocrine Tumors/drug therapy , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Animals , Humans , Neural Plate/drug effects , Neural Plate/metabolism , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
12.
Nat Commun ; 4: 1837, 2013.
Article in English | MEDLINE | ID: mdl-23673622

ABSTRACT

Calcium fluxes have been implicated in the specification of the vertebrate embryonic nervous system for some time, but how these fluxes are regulated and how they relate to the rest of the neural induction cascade is unknown. Here we describe Calfacilitin, a transmembrane calcium channel facilitator that increases calcium flux by generating a larger window current and slowing inactivation of the L-type CaV1.2 channel. Calfacilitin binds to this channel and is co-expressed with it in the embryo. Regulation of intracellular calcium by Calfacilitin is required for expression of the neural plate specifiers Geminin and Sox2 and for neural plate formation. Loss-of-function of Calfacilitin can be rescued by ionomycin, which increases intracellular calcium. Our results elucidate the role of calcium fluxes in early neural development and uncover a new factor in the modulation of calcium signalling.


Subject(s)
Calcium Channels/metabolism , Membrane Proteins/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Animals , Body Patterning/drug effects , Body Patterning/genetics , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Chick Embryo , Geminin/metabolism , Gene Expression Regulation, Developmental/drug effects , Germ Layers/cytology , Germ Layers/drug effects , Germ Layers/metabolism , HEK293 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Proteins/genetics , Molecular Sequence Data , Morpholinos/pharmacology , Neural Plate/drug effects , Quail
13.
PLoS One ; 8(5): e64368, 2013.
Article in English | MEDLINE | ID: mdl-23691206

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by abnormal polyglutamine expansion in the amino-terminal end of the huntingtin protein (Htt) and characterized by progressive striatal and cortical pathology. Previous reports have shown that Htt is essential for embryogenesis, and a recent study by our group revealed that the pathogenic form of Htt (mHtt) causes impairments in multiple stages of striatal development. In this study, we have examined whether HD-associated striatal developmental deficits are reflective of earlier maturational alterations occurring at the time of neurulation by assessing differential roles of Htt and mHtt during neural induction and early neurogenesis using an in vitro mouse embryonic stem cell (ESC) clonal assay system. We demonstrated that the loss of Htt in ESCs (KO ESCs) severely disrupts the specification of primitive and definitive neural stem cells (pNSCs, dNSCs, respectively) during the process of neural induction. In addition, clonally derived KO pNSCs and dNSCs displayed impaired proliferative potential, enhanced cell death and altered multi-lineage potential. Conversely, as observed in HD knock-in ESCs (Q111 ESCs), mHtt enhanced the number and size of pNSC clones, which exhibited enhanced proliferative potential and precocious neuronal differentiation. The transition from Q111 pNSCs to fibroblast growth factor 2 (FGF2)-responsive dNSCs was marked by potentiation in the number of dNSCs and altered proliferative potential. The multi-lineage potential of Q111 dNSCs was also enhanced with precocious neurogenesis and oligodendrocyte progenitor elaboration. The generation of Q111 epidermal growth factor (EGF)-responsive dNSCs was also compromised, whereas their multi-lineage potential was unaltered. These abnormalities in neural induction were associated with differential alterations in the expression profiles of Notch, Hes1 and Hes5. These cumulative observations indicate that Htt is required for multiple stages of neural induction, whereas mHtt enhances this process and promotes precocious neurogenesis and oligodendrocyte progenitor cell elaboration.


Subject(s)
Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Plate/embryology , Neurogenesis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Cell Proliferation/drug effects , Ectoderm/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endoderm/cytology , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Humans , Huntingtin Protein , Leukemia Inhibitory Factor/pharmacology , Mice , Neural Plate/cytology , Neural Plate/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Oligodendroglia/cytology , Receptors, Notch/metabolism , Signal Transduction/drug effects
14.
J Biol Chem ; 287(27): 22691-700, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22556407

ABSTRACT

Heparan sulfate (HS) has been implicated in regulating cell fate decisions during differentiation of embryonic stem cells (ESCs) into advanced cell types. However, the necessity and the underlying molecular mechanisms of HS in early cell lineage differentiation are still largely unknown. In this study, we examined the potential of EXT1(-/-) mouse ESCs (mESCs), that are deficient in HS, to differentiate into primary germ layer cells. We observed that EXT1(-/-) mESCs lost their differentiation competence and failed to differentiate into Pax6(+)-neural precursor cells and mesodermal cells. More detailed analyses highlighted the importance of HS for the induction of Brachyury(+) pan-mesoderm as well as normal gene expression associated with the dorso-ventral patterning of mesoderm. Examination of developmental cell signaling revealed that EXT1 ablation diminished FGF and BMP but not Wnt signaling. Furthermore, restoration of FGF and BMP signaling each partially rescued mesoderm differentiation defects. We further show that BMP4 is more prone to degradation in EXT1(-/-) mESCs culture medium compared with that of wild type cells. Therefore, our data reveal that HS stabilizes BMP ligand and thereby maintains the BMP signaling output required for normal mesoderm differentiation. In summary, our study demonstrates that HS is required for ESC pluripotency, in particular lineage specification into mesoderm through facilitation of FGF and BMP signaling.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Wnt Signaling Pathway/physiology , Animals , Anticoagulants/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Lineage/drug effects , Cell Lineage/physiology , Cells, Cultured , Culture Media/pharmacology , Ectoderm/cytology , Ectoderm/drug effects , Embryonic Stem Cells/drug effects , Fetal Proteins/genetics , Fetal Proteins/metabolism , Fibroblast Growth Factor 2/pharmacology , Heparin/pharmacology , Heparitin Sulfate/pharmacology , Mesoderm/cytology , Mesoderm/drug effects , Mice , Mice, Mutant Strains , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neural Plate/cytology , Neural Plate/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , RNA, Messenger/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway/drug effects
15.
Stem Cells ; 29(12): 1975-82, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21997878

ABSTRACT

Fibroblast growth factor (FGF) signaling and PAX6 transcription are required for neuroectoderm specification of human embryonic stem cells (hESCs). In this study, we asked how FGF signaling leads to PAX6 transcription and neuroectoderm specification from hESCs. Under a chemically defined medium, FGF inhibition blocked phosphorylation of extracellular signal-regulated kinase 1/2 (ERK 1/2) with a significant reduction of PAX6-expressing neuroepithelia, indicating that FGF regulates neural induction through ERK1/2 activation. Activation of FGF-ERK1/2 pathway was necessary for the activity of poly(ADP-ribose) polymerase-1 (PARP-1), a conserved nuclear protein catalyzing polymerization of ADP-ribose units. Pharmacological inhibition and genetic ablation of PARP-1 inhibited neural induction from hESCs, suggesting that FGF-ERK1/2 signal pathway regulates neuroectoderm specification through regulating PARP-1 activity. Furthermore, FGF-ERK1/2-PARP-1 cascade regulated the expression of PAX6, a transcription determinant of human neuroectoderm. Together, we propose that FGF regulates hESC neural specification through the ERK1/2-PARP-1 signaling pathway.


Subject(s)
Eye Proteins/metabolism , Fibroblast Growth Factors/metabolism , Homeodomain Proteins/metabolism , MAP Kinase Signaling System , Neural Plate/cytology , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Butadienes/pharmacology , Cell Differentiation , Cells, Cultured , Culture Media/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Enzyme Activation , Eye Proteins/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Neural Plate/drug effects , Neural Plate/metabolism , Nitriles/pharmacology , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Phenanthrenes/pharmacology , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Pyrroles/pharmacology , Repressor Proteins/genetics
16.
Bull Exp Biol Med ; 151(4): 502-5, 2011 Aug.
Article in English | MEDLINE | ID: mdl-22448377

ABSTRACT

We studied the effect of recombinant human activin A on induced neuroectoderm formation in colonies of human parthenogenetic SC in the absence of feeder cells. It was found that pretreatment of human parthenogenetic SC with activin A suppressed subsequent neural induction. Activin A in a concentration of 10 ng/ml significantly decreased transcriptional activity of genes required for neuroectoderm formation. At the same time, activin A in a concentration of 20 ng/ml increased the expression of pluripotency genes and completely inhibited the formation of structures in vitro reproducing the neural tube of the developing embryo. These findings attest to prolonged effect of activin A as an inhibitor of neuroectodermal differentiation.


Subject(s)
Activins/pharmacology , Neural Plate/drug effects , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Mice , Parthenogenesis , Recombinant Proteins/pharmacology , Transcription, Genetic/drug effects
17.
Indian J Exp Biol ; 48(4): 346-53, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20726332

ABSTRACT

Gastrulation is a fundamental process that results in formation of the three germ layers in an embryo. It involves highly coordinated cell migration. Cell to cell communication through cell surface and the surrounding molecular environment governs cell migration. In the present work, cell surface features, which are indicative of the migratory status of a cell, of an early gastrulating chick embryo were studied using scanning electron microscopy. The distinct ultrastructural features of cells located in the various regions of the epiblast are described. Differences in the surface features of cells from distinct embryonic regions indicate differences in their migratory capacities. Further, the dynamic nature of these cell surface features by their response to altered fibroblast growth factor (FGF) signaling, experimentally created by using either excess FGF or inhibition of FGF signaling are demonstrated.


Subject(s)
Fibroblast Growth Factors/metabolism , Gastrulation/drug effects , Neural Plate/ultrastructure , Organizers, Embryonic/ultrastructure , Primitive Streak/ultrastructure , Signal Transduction/drug effects , Animals , Chick Embryo , Fibroblast Growth Factor 2/pharmacology , Humans , In Vitro Techniques , Microscopy, Electron, Scanning , Neural Plate/drug effects , Neural Plate/embryology , Neural Plate/metabolism , Organizers, Embryonic/drug effects , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , Primitive Streak/drug effects , Primitive Streak/embryology , Primitive Streak/metabolism , Recombinant Proteins/pharmacology , Suramin/pharmacology
18.
Stem Cells ; 28(10): 1772-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20715182

ABSTRACT

Formation of the neural plate is an intricate process in early mammalian embryonic development mediated by cells of the inner cell mass and involving a series of steps, including development of the epiblast. Here, we report on the creation of an embryonic stem (ES) cell-based system to isolate and identify neural induction intermediates with characteristics of epiblast cells and neural plate. We demonstrate that neural commitment requires prior differentiation of ES cells into epiblast cells that are indistinguishable from those derived from natural embryos. We also demonstrate that epiblast cells can be isolated and cultured as epiblast stem cell lines. Fgf signaling is shown to be required for the differentiation of ES cells into these epiblast cells. Fgf2, widely used for maintenance of both human ES cells and epiblast stem cells, inhibits formation of early neural cells by epiblast intermediates in a dose-dependent manner and is sufficient to promote transient self-renewal of epiblast stem cells. In contrast, Fgf8, the endogenous embryonic neural inducer, fails to promote epiblast self-renewal, but rather promotes more homogenous neural induction with transient self-renewal of early neural cells. Removal of Fgf signaling entirely from epiblast cells promotes rapid neural induction and subsequent neurogenesis. We conclude that Fgf signaling plays different roles during the differentiation of ES cells, with an initial requirement in epiblast formation and a subsequent role in self-renewal. Fgf2 and Fgf8 thus stimulate self-renewal in different cell types.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 8/pharmacology , Germ Layers/cytology , Germ Layers/drug effects , Humans , Male , Mice , Neural Plate/cytology , Neural Plate/drug effects , Neurogenesis/drug effects , Neurogenesis/genetics , Signal Transduction/drug effects
19.
Stem Cells ; 28(9): 1498-509, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20665854

ABSTRACT

Embryonic stem (ES) cells differentiate spontaneously toward a neuroectodermal fate in serum-free, adherent monocultures. Here, we show that this spontaneous neural fate requires retinoic acid (RA) synthesis. We monitor ES cells containing reporter genes for markers of the early neural plate as well as the primitive streak and its progeny to determine the cell fates induced when RA signaling is perturbed. We demonstrate that the spontaneous neural commitment of mouse ES cells requires endogenous RA production from vitamin A (vitA) in the medium. Formation of neural progenitors is inhibited by removing vitA from the medium, by inhibiting the enzymes that catalyze the synthesis of RA, or by inhibiting RA receptors. We show that subnanomolar concentrations of RA restore neuroectodermal differentiation when RA synthesis is blocked. We demonstrate that a neural to mesodermal fate change occurring when RA signaling is inhibited is dependent on Nodal-, Wnt-, and fibroblast growth factor-signaling. We show that Nodal suppresses neural development in a Wnt-dependent manner and that Wnt-mediated inhibition of neural development is reversed by inhibition of Nodal signaling. Together, our results show that neural induction in ES cells requires RA at subnanomolar levels to suppress Nodal signaling and suggest that the mechanism by which Wnt signaling suppresses neural development is through facilitation of Nodal signaling.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Neural Plate/metabolism , Neurons/metabolism , Nodal Protein/metabolism , Signal Transduction , Tretinoin/metabolism , Wnt Proteins/metabolism , Acyclic Monoterpenes , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Disulfiram/pharmacology , Embryonic Stem Cells/drug effects , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Mesoderm/cytology , Mesoderm/metabolism , Mice , Monoterpenes/pharmacology , Naphthalenes , Neural Plate/cytology , Neural Plate/drug effects , Neurons/drug effects , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Vitamin A/metabolism
20.
Exp Cell Res ; 316(7): 1148-58, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20171210

ABSTRACT

In this study we demonstrated that neural rosettes derived from human ES cells can give rise either to neural crest precursors, following expansion in presence of bFGF and EGF, or to dopaminergic precursors after exposure to ventralizing factors Shh and FGF8. Both regionalised precursors are capable of extensive proliferation and differentiation towards the corresponding terminally differentiated cell types. In particular, peripheral neurons, cartilage, bone, smooth muscle cells and also pigmented cells were obtained from neural crest precursors while tyrosine hydroxylase and Nurr1 positive dopaminergic neurons were derived from FGF8 and Shh primed rosette cells. Gene expression and immunocytochemistry analyses confirmed the expression of dorsal and neural crest genes such as Sox10, Slug, p75, FoxD3, Pax7 in neural precursors from bFGF-EGF exposed rosettes. By contrast, priming of rosettes with FGF8 and Shh induced the expression of dopaminergic markers Engrailed1, Pax2, Pitx3, floor plate marker FoxA2 and radial glia markers Blbp and Glast, the latter in agreement with the origin of dopaminergic precursors from floor plate radial glia. Moreover, in vivo transplant of proliferating Shh/FGF8 primed precursors in parkinsonian rats demonstrated engraftment and terminal dopaminergic differentiation. In conclusion, we demonstrated the derivation of long-term self-renewing precursors of selected regional identity as potential cell reservoirs for cell therapy applications, such as CNS degenerative diseases, or for the development of toxicological tests.


Subject(s)
Cell Differentiation/drug effects , Central Nervous System/drug effects , Fibroblast Growth Factor 8/pharmacology , Hedgehog Proteins/pharmacology , Neural Plate/drug effects , Stem Cells/drug effects , Animals , Body Patterning/drug effects , Cell Culture Techniques , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Central Nervous System/embryology , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Neural Plate/embryology , Neurons/drug effects , Neurons/physiology , Neurons/transplantation , Rats , Rats, Sprague-Dawley , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Spheroids, Cellular/transplantation , Stem Cells/physiology , Time Factors , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...