Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nucleic Acids Res ; 45(12): 7118-7136, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28449087

ABSTRACT

Cytosine modifications diversify and structure the genome thereby controlling proper development and differentiation. Here, we focus on the interplay of the 5-methylcytosine reader Mbd1 and modifier Tet1 by analyzing their dynamic subcellular localization and the formation of the Tet oxidation product 5-hydroxymethylcytosine in mammalian cells. Our results demonstrate that Mbd1 enhances Tet1-mediated 5-methylcytosine oxidation. We show that this is due to enhancing the localization of Tet1, but not of Tet2 and Tet3 at heterochromatic DNA. We find that the recruitment of Tet1 and concomitantly its catalytic activity eventually leads to the displacement of Mbd1 from methylated DNA. Finally, we demonstrate that increased Tet1 heterochromatin localization and 5-methylcytosine oxidation are dependent on the CXXC3 domain of Mbd1, which recognizes unmethylated CpG dinucleotides. The Mbd1 CXXC3 domain deletion isoform, which retains only binding to methylated CpGs, on the other hand, blocks Tet1-mediated 5-methylcytosine to 5-hydroxymethylcytosine conversion, indicating opposite biological effects of Mbd1 isoforms. Our study provides new insights on how cytosine modifications, their modifiers and readers cross-regulate themselves.


Subject(s)
CpG Islands , DNA-Binding Proteins/genetics , DNA/metabolism , Gene Expression Regulation , Heterochromatin/metabolism , Mixed Function Oxygenases/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Cell Line , DNA/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Heterochromatin/chemistry , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mixed Function Oxygenases/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Oxidation-Reduction , Protein Domains , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Red Fluorescent Protein
2.
Biochem Biophys Res Commun ; 478(2): 573-9, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27470587

ABSTRACT

Podocalyxin (PODXL) is a highly glycosylated and sialylated transmembrane protein that is up-regulated in various types of tumors and whose expression levels positively correlate with tumor grade. We previously found Podxl to be highly expressed in murine tumorigenic neural stem/progenitor cells (NSPs). Here we investigated the effects of elevated Podxl levels in these cells. NSPs overexpressing Podxl did not form brain tumors upon intracranial transplantations, indicating that high levels of this gene alone are not sufficient for tumor initiation. However, Podxl overexpression had a positive effect on cell number, sphere formation and cell viability, indicating that it might in this way contribute to the development and/or maintenance of tumors. Proteome analyses of Podxl-overexpressing and control NSPs revealed increased levels of Annexin A2 (ANXA2). We also found increased transcript levels, indicating that PODXL stimulates expression of the Anxa2 gene. Lack of Anxa2 in Podxl-overexpressing NSPs resulted in reduced viability of these cells, suggesting that PODXL-mediated pro-survival effects can at least in part be explained by increased ANXA2 levels. Finally, our data indicate that Podxl overexpression activates the MAP kinase (MAPK) pathway which in turn up-regulates Anxa2 expression. Our data indicate a novel molecular connection between PODXL and ANXA2: both exert pro-survival effects in NSPs, and PODXL positively regulates ANXA2 expression through the MAPK pathway.


Subject(s)
Annexin A2/genetics , Cell Survival , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Sialoglycoproteins/genetics , Up-Regulation , Animals , Annexin A2/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line , Cell Proliferation , Cells, Cultured , Down-Regulation , MAP Kinase Signaling System , Mice, Inbred C57BL , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Neural Stem Cells/cytology , Neural Stem Cells/pathology , Sialoglycoproteins/metabolism , Transcriptional Activation
3.
Hum Mol Genet ; 21(8): 1673-80, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22186023

ABSTRACT

Rett syndrome (RTT) is caused by loss-of-function mutations in the X-linked gene MECP2 coding for methyl CpG-binding protein 2 (MeCP2). This protein can act as transcriptional repressor, and we showed in a previous study that glucocorticoid-inducible genes are up-regulated in an RTT mouse model and that these genes are direct MeCP2 targets. Here, we report that pharmacological intervention with the glucocorticoid system has an impact on the symptoms and lifespan in an RTT mouse model. Our data support a functional implication of the stress hormone system in RTT and suggest this hormone system as potential therapeutic target.


Subject(s)
Corticosterone/pharmacology , Glucocorticoids/metabolism , Mifepristone/pharmacology , Receptors, Glucocorticoid/metabolism , Rett Syndrome/physiopathology , Animals , Corticosterone/administration & dosage , Corticosterone/blood , Disease Models, Animal , Female , Gene Expression Regulation , Immediate-Early Proteins/genetics , Life Expectancy , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mifepristone/metabolism , Motor Activity/drug effects , Protein Serine-Threonine Kinases/genetics , Receptors, Glucocorticoid/antagonists & inhibitors , Rett Syndrome/genetics , Rotarod Performance Test , Tacrolimus Binding Proteins/genetics
4.
J Neurosci ; 32(15): 5151-64, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496561

ABSTRACT

Ischemic stroke causes transient increase of neural stem and progenitor cell (NSPC) proliferation in the subventricular zone (SVZ), and migration of newly formed neuroblasts toward the damaged area where they mature to striatal neurons. The molecular mechanisms regulating this plastic response, probably involved in structural reorganization and functional recovery, are poorly understood. The adaptor protein LNK suppresses hematopoietic stem cell self-renewal, but its presence and role in the brain are poorly understood. Here we demonstrate that LNK is expressed in NSPCs in the adult mouse and human SVZ. Lnk(-/-) mice exhibited increased NSPC proliferation after stroke, but not in intact brain or following status epilepticus. Deletion of Lnk caused increased NSPC proliferation while overexpression decreased mitotic activity of these cells in vitro. We found that Lnk expression after stroke increased in SVZ through the transcription factors STAT1/3. LNK attenuated insulin-like growth factor 1 signaling by inhibition of AKT phosphorylation, resulting in reduced NSPC proliferation. Our findings identify LNK as a stroke-specific, endogenous negative regulator of NSPC proliferation, and suggest that LNK signaling is a novel mechanism influencing plastic responses in postischemic brain.


Subject(s)
Brain Ischemia/pathology , Brain/cytology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Neural Stem Cells/physiology , Stroke/pathology , Adaptor Proteins, Signal Transducing , Animals , Antimetabolites , Bromodeoxyuridine , Cell Proliferation , Cell Survival , Cells, Cultured , Chromatin Immunoprecipitation , Electroporation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Male , Membrane Proteins , Mice , Mice, Knockout , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Real-Time Polymerase Chain Reaction , Recovery of Function , Retroviridae/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Transcription Factors/metabolism , Transfection/methods
5.
Stem Cell Reports ; 18(11): 2240-2253, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37922914

ABSTRACT

In early vertebrate development, organizer regions-groups of cells that signal to and thereby influence neighboring cells by secreted morphogens-play pivotal roles in the establishment and maintenance of cell identities within defined tissue territories. The midbrain-hindbrain organizer drives regionalization of neural tissue into midbrain and hindbrain territories with fibroblast growth factor 8 (FGF8) acting as a key morphogen. This organizer has been extensively studied in chicken, mouse, and zebrafish. Here, we demonstrate the enrichment of FGF8-expressing cells from human pluripotent stem cells (hPSCs), cultured as attached embryoid bodies using antibodies that recognize "Similar Expression to Fgf" (SEF) and Frizzled proteins. The arrangement of cells in embryoid body subsets of these cultures and the gene expression profile of the FGF8-expressing population show certain similarities to the midbrain-hindbrain organizer in animal models. In the embryonic chick brain, the enriched cell population induces formation of midbrain structures, consistent with FGF8-organizing capability.


Subject(s)
Homeodomain Proteins , Pluripotent Stem Cells , Humans , Animals , Mice , Homeodomain Proteins/metabolism , Zebrafish/metabolism , Fibroblast Growth Factor 8/genetics , Chickens/metabolism , Mesencephalon/metabolism , Pluripotent Stem Cells/metabolism , Gene Expression Regulation, Developmental , Fibroblast Growth Factors/metabolism , Body Patterning
6.
Front Cell Dev Biol ; 10: 941493, 2022.
Article in English | MEDLINE | ID: mdl-36172281

ABSTRACT

Rett syndrome is a human intellectual disability disorder that is associated with mutations in the X-linked MECP2 gene. The epigenetic reader MeCP2 binds to methylated cytosines on the DNA and regulates chromatin organization. We have shown previously that MECP2 Rett syndrome missense mutations are impaired in chromatin binding and heterochromatin reorganization. Here, we performed a proteomics analysis of post-translational modifications of MeCP2 isolated from adult mouse brain. We show that MeCP2 carries various post-translational modifications, among them phosphorylation on S80 and S421, which lead to minor changes in either heterochromatin binding kinetics or clustering. We found that MeCP2 is (di)methylated on several arginines and that this modification alters heterochromatin organization. Interestingly, we identified the Rett syndrome mutation site R106 as a dimethylation site. In addition, co-expression of protein arginine methyltransferases (PRMT)1 and PRMT6 lead to a decrease of heterochromatin clustering. Altogether, we identified and validated novel modifications of MeCP2 in the brain and show that these can modulate its ability to bind as well as reorganize heterochromatin, which may play a role in the pathology of Rett syndrome.

7.
Glia ; 59(1): 68-81, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21046556

ABSTRACT

In contrast to ependymal cells located above the subventricular zone (SVZ) of the adult lateral ventricle wall (LVW), adult spinal cord (SC) ependymal cells possess certain neural stem cell characteristics. The molecular basis of this difference is unknown. In this study, antibodies against multiple cell surface markers were applied to isolate pure populations of SC and LVW ependymal cells, which allowed a direct comparison of their in vitro behavior and in vivo gene expression profile. Isolated CD133(+)/CD24(+)/CD45(-)/CD34(-) ependymal cells from the SC displayed in vitro self-renewal and differentiation capacity, whereas those from the LVW did not. SC ependymal cells showed a higher expression of several genes involved in cell division, cell cycle regulation, and chromosome stability, which is consistent with a long-term self-renewal capacity, and shared certain transcripts with neural stem cells of the embryonic forebrain. They also expressed several retinoic acid (RA)-regulated genes and responded to RA exposure. LVW ependymal cells showed higher transcript levels of many genes regulated by transforming growth factor-ß family members. Among them were Dlx2, Id2, Hey1, which together with Foxg1 could explain their potential to turn into neuroblasts under certain environmental conditions.


Subject(s)
Antigens, CD/metabolism , CD24 Antigen/metabolism , Ependyma/cytology , Gene Expression , Glycoproteins/metabolism , Lateral Ventricles/cytology , Peptides/metabolism , Spinal Cord/cytology , AC133 Antigen , Animals , Cell Differentiation , Cells, Cultured , Ependyma/metabolism , Flow Cytometry , Immunohistochemistry , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism
8.
Nucleic Acids Res ; 37(11): e82, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19443447

ABSTRACT

The prediction of transcription factor binding sites in genomic sequences is in principle very useful to identify upstream regulatory factors. However, when applying this concept to genomes of multicellular organisms such as mammals, one has to deal with a large number of false positive predictions since many transcription factor genes are only expressed in specific tissues or cell types. We developed TS-REX, a database/software system that supports the analysis of tissue and cell type-specific transcription factor-gene networks based on expressed sequence tag abundance of transcription factor-encoding genes in UniGene EST libraries. The use of expression levels of transcription factor-encoding genes according to hierarchical anatomical classifications covering different tissues and cell types makes it possible to filter out irrelevant binding site predictions and to identify candidates of potential functional importance for further experimental testing. TS-REX covers ESTs from H. sapiens and M. musculus, and allows the characterization of both presence and specificity of transcription factors in user-specified tissues or cell types. The software allows users to interactively visualize transcription factor-gene networks, as well as to export data for further processing. TS-REX was applied to predict regulators of Polycomb group genes in six human tumor tissues and in human embryonic stem cells.


Subject(s)
Databases, Genetic , Gene Regulatory Networks , Software , Transcription Factors/metabolism , Animals , Binding Sites , Cell Line, Tumor , Embryonic Stem Cells/metabolism , Expressed Sequence Tags , Gene Expression Regulation , Gene Library , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Polycomb-Group Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
9.
Cancer Res ; 67(12): 5727-36, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575139

ABSTRACT

Human brain tumor stem cells have been enriched using antibodies against the surface protein CD133. An antibody recognizing CD133 also served to isolate normal neural stem cells from fetal human brain, suggesting a possible lineage relationship between normal neural and brain tumor stem cells. Whether CD133-positive brain tumor stem cells can be derived from CD133-positive neural stem or progenitor cells still requires direct experimental evidence, and an important step toward such investigations is the identification and characterization of normal CD133-presenting cells in neurogenic regions of the embryonic and adult brain. Here, we present evidence that CD133 is a marker for embryonic neural stem cells, an intermediate radial glial/ependymal cell type in the early postnatal stage, and for ependymal cells in the adult brain, but not for neurogenic astrocytes in the adult subventricular zone. Our findings suggest two principal possibilities for the origin of brain tumor stem cells: a derivation from CD133-expressing cells, which are normally not present in the adult brain (embryonic neural stem cells and an early postnatal intermediate radial glial/ependymal cell type), or from CD133-positive ependymal cells in the adult brain, which are, however, generally regarded as postmitotic. Alternatively, brain tumor stem cells could be derived from proliferative but CD133-negative neurogenic astrocytes in the adult brain. In the latter case, brain tumor development would involve the production of CD133.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Embryonic Stem Cells/metabolism , Ependyma/metabolism , Glioblastoma/metabolism , Glycoproteins/metabolism , Peptides/metabolism , Prosencephalon/metabolism , AC133 Antigen , Adult , Animals , Astrocytes/metabolism , Blotting, Western , Ependyma/cytology , Epithelial Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neuroglia/metabolism , Prosencephalon/cytology , Prosencephalon/growth & development
10.
Nat Commun ; 10(1): 2966, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31273213

ABSTRACT

Mutations in genes encoding components of BAF (BRG1/BRM-associated factor) chromatin remodeling complexes cause neurodevelopmental disorders and tumors. The mechanisms leading to the development of these two disease entities alone or in combination remain unclear. We generated mice with a heterozygous nervous system-specific partial loss-of-function mutation in a BAF core component gene, Smarcb1. These Smarcb1 mutant mice show various brain midline abnormalities that are also found in individuals with Coffin-Siris syndrome (CSS) caused by SMARCB1, SMARCE1, and ARID1B mutations and in SMARCB1-related intellectual disability (ID) with choroid plexus hyperplasia (CPH). Analyses of the Smarcb1 mutant animals indicate that one prominent midline abnormality, corpus callosum agenesis, is due to midline glia aberrations. Our results establish a novel role of Smarcb1 in the development of the brain midline and have important clinical implications for BAF complex-related ID/neurodevelopmental disorders.


Subject(s)
Abnormalities, Multiple/genetics , Agenesis of Corpus Callosum/genetics , Corpus Callosum/growth & development , Face/abnormalities , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , Micrognathism/genetics , Neck/abnormalities , SMARCB1 Protein/genetics , Abnormalities, Multiple/diagnostic imaging , Agenesis of Corpus Callosum/diagnostic imaging , Agenesis of Corpus Callosum/pathology , Alleles , Animals , Child , Child, Preschool , Corpus Callosum/cytology , Corpus Callosum/diagnostic imaging , Disease Models, Animal , Embryo, Mammalian , Face/diagnostic imaging , Female , Hand Deformities, Congenital/diagnostic imaging , Humans , Infant , Intellectual Disability/diagnostic imaging , Loss of Function Mutation , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Micrognathism/diagnostic imaging , Neck/diagnostic imaging , Neuroglia/pathology , Primary Cell Culture
11.
PLoS Comput Biol ; 2(9): e123, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16978048

ABSTRACT

Recent ChIP experiments of human and mouse embryonic stem cells have elucidated the architecture of the transcriptional regulatory circuitry responsible for cell determination, which involves the transcription factors OCT4, SOX2, and NANOG. In addition to regulating each other through feedback loops, these genes also regulate downstream target genes involved in the maintenance and differentiation of embryonic stem cells. A search for the OCT4-SOX2-NANOG network motif in other species reveals that it is unique to mammals. With a kinetic modeling approach, we ascribe function to the observed OCT4-SOX2-NANOG network by making plausible assumptions about the interactions between the transcription factors at the gene promoter binding sites and RNA polymerase (RNAP), at each of the three genes as well as at the target genes. We identify a bistable switch in the network, which arises due to several positive feedback loops, and is switched on/off by input environmental signals. The switch stabilizes the expression levels of the three genes, and through their regulatory roles on the downstream target genes, leads to a binary decision: when OCT4, SOX2, and NANOG are expressed and the switch is on, the self-renewal genes are on and the differentiation genes are off. The opposite holds when the switch is off. The model is extremely robust to parameter changes. In addition to providing a self-consistent picture of the transcriptional circuit, the model generates several predictions. Increasing the binding strength of NANOG to OCT4 and SOX2, or increasing its basal transcriptional rate, leads to an irreversible bistable switch: the switch remains on even when the activating signal is removed. Hence, the stem cell can be manipulated to be self-renewing without the requirement of input signals. We also suggest tests that could discriminate between a variety of feedforward regulation architectures of the target genes by OCT4, SOX2, and NANOG.


Subject(s)
Embryonic Stem Cells/metabolism , Transcription, Genetic/genetics , Animals , Computational Biology , Computer Simulation , Gene Regulatory Networks , High Mobility Group Proteins/genetics , Homeodomain Proteins/genetics , Humans , Models, Biological , Octamer Transcription Factor-3/genetics , Phylogeny
12.
Oncotarget ; 7(36): 58203-58217, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27533460

ABSTRACT

Bmi1 was originally identified as a gene that contributes to the development of mouse lymphoma by inhibiting MYC-induced apoptosis through repression of Ink4a and Arf. It codes for the Polycomb group protein BMI-1 and acts primarily as a transcriptional repressor via chromatin modifications. Although it binds to a large number of genomic regions, the direct BMI-1 target genes described so far do not explain the full spectrum of BMI-1-mediated effects. Here we identify the putative tumor suppressor gene EphA7 as a novel direct BMI-1 target in neural cells and lymphocytes. EphA7 silencing has been reported in several different human tumor types including lymphomas, and our data suggest BMI1 overexpression as a novel mechanism leading to EphA7 inactivation via H3K27 trimethylation and DNA methylation.


Subject(s)
Gene Expression Regulation , Genes, Tumor Suppressor , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, EphA7/genetics , Animals , B-Lymphocytes , Cell Culture Techniques/methods , Cell Nucleus/metabolism , Cell Proliferation/physiology , Cells, Cultured , Cerebellum/anatomy & histology , Cerebellum/metabolism , DNA Methylation/physiology , Down-Regulation , Histones/metabolism , Immunohistochemistry , Ki-67 Antigen/metabolism , Lateral Ventricles/anatomy & histology , Lateral Ventricles/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Neural Stem Cells , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , Receptor, EphA7/metabolism , Spleen/cytology , Transduction, Genetic , Up-Regulation
13.
J Neurosci ; 24(26): 5982-6002, 2004 Jun 30.
Article in English | MEDLINE | ID: mdl-15229246

ABSTRACT

The molecular changes underlying neural progenitor differentiation are essentially unknown. We applied cDNA microarrays with 13,627 clones to measure dynamic gene expression changes during the in vitro differentiation of neural progenitor cells that were isolated from the subventricular zone of postnatal day 7 mice and grown in vitro as neurospheres. In two experimental series in which we withdrew epidermal growth factor and added the neurotrophins Neurotrophin-4 or BDNF, four time points were investigated: undifferentiated cells grown as neurospheres, and cells 24, 48, and 96 hr after differentiation. Expression changes of selected genes were confirmed by semiquantitative RT-PCR. Ten different groups of gene expression dynamics obtained by cluster analysis are described. To correlate selected gene expression changes to the localization of respective proteins, we performed immunostainings of cultured neurospheres and of brain sections from adult mice. Our results provide new insights into the genetic program of neural progenitor differentiation and give strong hints to as yet unknown cellular communications within the adult subventricular zone stem cell niche.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Lateral Ventricles/growth & development , Nerve Tissue Proteins/biosynthesis , Neurons/cytology , Stem Cells/cytology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation/drug effects , Cell Division , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , DNA, Complementary/genetics , Gene Expression Regulation, Developmental/drug effects , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spheroids, Cellular/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
14.
Eur J Cell Biol ; 84(2-3): 123-35, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15819395

ABSTRACT

Epigenetics is a term that has changed its meaning with the increasing biological knowledge on developmental processes. However, its current application to stem cell biology is often imprecise and is conceptually problematic. This article addresses two different subjects, the definition of epigenetics and chromatin states of stem and differentiated cells. We describe mechanisms that regulate chromatin changes and provide an overview of chromatin states of stem and differentiated cells. Moreover, a modification of the current epigenetics definition is proposed that is not restricted by the heritability of gene expression throughout cell divisions and excludes translational gene expression control.


Subject(s)
Chromatin/physiology , Epigenesis, Genetic/physiology , Stem Cells/physiology , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly/physiology , Chromosomal Proteins, Non-Histone/physiology , DNA Methylation , Gene Expression Regulation/physiology , Histones/physiology , Humans , RNA, Untranslated/physiology
15.
BMC Genomics ; 4(1): 1, 2003 Jan 16.
Article in English | MEDLINE | ID: mdl-12529184

ABSTRACT

BACKGROUND: Methylation at CpG dinucleotides in genomic DNA is a fundamental epigenetic mechanism of gene expression control in vertebrates. Proteins with a methyl-CpG-binding domain (MBD) can bind to single methylated CpGs and most of them are involved in transcription control. So far, five vertebrate MBD proteins have been described as MBD family members: MBD1, MBD2, MBD3, MBD4 and MECP2. RESULTS: We performed database searches for new proteins containing an MBD and identified six amino acid sequences which are different from the previously described ones. Here we present a comparison of their MBD sequences, additional protein motifs and the expression of the encoding genes. A calculated unrooted dendrogram indicates the existence of at least four different groups of MBDs within these proteins. Two of these polypeptides, KIAA1461 and KIAA1887, were only present as predicted amino acid sequences based on a partial human cDNA. We investigated their expression by Northern blot analysis and found transcripts of ~8 kb and ~5 kb respectively, in all eight normal tissues studied. CONCLUSIONS: Eleven polypeptides with a MBD could be identified in mouse and man. The analysis of protein domains suggests a role in transcriptional regulation for most of them. The knowledge of additional existing MBD proteins and their expression pattern is important in the context of Rett syndrome.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Cells, Cultured , CpG Islands , DNA Methylation , DNA-Binding Proteins/genetics , Humans , Mice , Molecular Sequence Data , Phylogeny , Protein Structure, Tertiary , Sequence Alignment , Transcription, Genetic
16.
Invest Ophthalmol Vis Sci ; 43(9): 2825-33, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12202498

ABSTRACT

PURPOSE: Mutations in the NDP gene give rise to a variety of eye diseases, including classic Norrie disease (ND), X-linked exudative vitreoretinopathy (EVRX), retinal telangiectasis (Coats disease), and advanced retinopathy of prematurity (ROP). The gene product is a cystine-knot-containing extracellular signaling molecule of unknown function. In the current study, gene expression was determined in a mouse model of ND, to unravel disease-associated mechanisms at the molecular level. METHODS: Gene transcription in the eyes of 2-year-old Ndp knockout mice was compared with that in the eyes of age-matched wild-type control animals, by means of cDNA subtraction and microarrays. Clones (n = 3072) from the cDNA subtraction libraries were spotted onto glass slides and hybridized with fluorescently labeled RNA-derived targets. More than 230 differentially expressed clones were sequenced, and their expression patterns were verified by virtual Northern blot analysis. RESULTS: Numerous gene transcripts that are absent or downregulated in the eye of Ndp knockout mice are photoreceptor cell specific. In younger Ndp knockout mice (up to 1 year old), however, all these transcripts were found to be expressed at normal levels. CONCLUSIONS: The identification of numerous photoreceptor cell-specific transcripts with a reduced expression in 2-year-old, but not in young, Ndp knockout mice indicates that normal gene expression in these light-sensitive cells of mutant mice is established and maintained over a long period and that rods and cones are affected relatively late in the mouse model of ND. Obviously, the absence of the Ndp gene product is not compatible with long-term survival of photoreceptor cells in the mouse.


Subject(s)
Blindness/genetics , Photoreceptor Cells, Vertebrate/pathology , Retinal Detachment/genetics , X Chromosome/genetics , Animals , Blindness/congenital , Blindness/pathology , Blotting, Northern , DNA, Complementary/analysis , Deafness/genetics , Disease Models, Animal , Disease Progression , Eye Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation , Intellectual Disability/genetics , Mice , Mice, Knockout/genetics , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Retinal Detachment/pathology , Sequence Analysis, DNA
17.
J Transl Med ; 2(1): 32, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15383145

ABSTRACT

Recently BRCA1 has been implicated in the regulation of gene expression from the X chromosome. In this study the influence of BRCA1 on expression of X chromosome genes was investigated. Complementary DNA microarrays were used to compare the expression levels of X chromosome genes in 18 BRCA1-associated ovarian cancers to those of the 13 "BRCA1-like" and 14 "BRCA2-like" sporadic tumors (as defined by previously reported expression profiling). Significance was determined using parametric statistics with P < 0.005 as a cutoff. Forty of 178 total X-chromosome transcripts were differentially expressed between the BRCA1-associated tumors and sporadic cancers with a BRCA2-like molecular profile. Thirty of these 40 genes showed higher mean expression in the BRCA1-associated samples including all 11 transcripts that mapped to Xp11. In contrast, four of 178 total X chromosome transcripts showed significant differential expression between BRCA1-associated and sporadic tumors with a BRCA1-like molecular profile. All four mapped to Xp11 and showed higher mean expression in BRCA1-associated tumors. Re-expression of BRCA1 in HCC1937 BRCA1-deficient breast cancer cell resulted in the repression of 21 transcripts. Eleven of the 21 (54.5%) transcripts mapped to Xp11. However, there was no significant overlap between these Xp11 genes and those found to be differentially expressed between BRCA1-associated and sporadic ovarian cancer samples. These results demonstrate that BRCA1 mediates the repression of several X chromosome genes, many of which map to the Xp11 locus.

18.
Stem Cells Dev ; 23(16): 1844-57, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24809660

ABSTRACT

True tendon regeneration in human patients remains a vision of musculoskeletal therapies. In comparison to other mesenchymal lineages the biology of tenogenic differentiation is barely understood. Specifically, easy and efficient protocols are lacking that might enable tendon cell and tissue differentiation based on adult (stem) cell sources. In the murine mesenchymal progenitor cell line C3H10T½, overexpression of the growth factor bone morphogenetic protein 2 (BMP2) and a constitutively active transcription factor, Smad8 L+MH2, mediates tendon cell differentiation in vitro and the formation of tendon-like tissue in vivo. We hypothesized that during this differentiation secreted factors involved in extracellular matrix formation exert a major impact on tendon development. Gene expression analyses revealed four genes encoding secreted factors that are notably upregulated: periostin, C-type lectin domain family 3 (member b), RNase A4, and follistatin-like 1. These factors have not previously been implicated in tendon biology. Among these, periostin showed a specific expression in tenocytes of adult mouse Achilles tendon and in chondrocytes within the nonmineralized fibrocartilage zone of the enthesis with the calcaneus. Overexpression of periostin alone or in combination with constitutively active BMP receptor type in human mesenchymal stem cells and subsequent implantation into ectopic sites in mice demonstrated a reproducible moderate tenogenic capacity that has not been described before. Therefore, periostin may belong to the factors contributing to the development of tenogenic tissue.


Subject(s)
Achilles Tendon/physiopathology , Cell Adhesion Molecules/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Bone and Bones/metabolism , Cell Adhesion Molecules/physiology , Cell Differentiation , Endoribonucleases/genetics , Endoribonucleases/metabolism , Female , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Gene Expression , HEK293 Cells , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice, Nude , Regeneration , Up-Regulation
19.
Diabetes ; 62(8): 2834-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23610061

ABSTRACT

Precise regulation of ß-cell function is crucial for maintaining blood glucose homeostasis. Pax6 is an essential regulator of ß-cell-specific factors like insulin and Glut2. Studies in the developing eye suggest that Pax6 interacts with Mitf to regulate pigment cell differentiation. Here, we show that Mitf, like Pax6, is expressed in all pancreatic endocrine cells during mouse postnatal development and in the adult islet. A Mitf loss-of-function mutation results in improved glucose tolerance and enhanced insulin secretion but no increase in ß-cell mass in adult mice. Mutant ß-cells secrete more insulin in response to glucose than wild-type cells, suggesting that Mitf is involved in regulating ß-cell function. In fact, the transcription of genes critical for maintaining glucose homeostasis (insulin and Glut2) and ß-cell formation and function (Pax4 and Pax6) is significantly upregulated in Mitf mutant islets. The increased Pax6 expression may cause the improved ß-cell function observed in Mitf mutant animals, as it activates insulin and Glut2 transcription. Chromatin immunoprecipitation analysis shows that Mitf binds to Pax4 and Pax6 regulatory regions, suggesting that Mitf represses their transcription in wild-type ß-cells. We demonstrate that Mitf directly regulates Pax6 transcription and controls ß-cell function.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Mutation , Animals , Blood Glucose/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Microphthalmia-Associated Transcription Factor/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcriptional Activation
20.
Cancer Res ; 72(13): 3381-92, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22719073

ABSTRACT

Although brain tumors are classified and treated based upon their histology, the molecular factors involved in the development of various tumor types remain unknown. In this study, we show that the type and order of genetic events directs the development of gliomas, central nervous system primitive neuroectodermal tumors, and atypical teratoid/rhabdoid-like tumors from postnatal mouse neural stem/progenitor cells (NSC/NPC). We found that the overexpression of specific genes led to the development of these three different brain tumors from NSC/NPCs, and manipulation of the order of genetic events was able to convert one established tumor type into another. In addition, loss of the nuclear chromatin-remodeling factor SMARCB1 in rhabdoid tumors led to increased phosphorylation of eIF2α, a central cytoplasmic unfolded protein response (UPR) component, suggesting a role for the UPR in these tumors. Consistent with this, application of the proteasome inhibitor bortezomib led to an increase in apoptosis of human cells with reduced SMARCB1 levels. Taken together, our findings indicate that the order of genetic events determines the phenotypes of brain tumors derived from a common precursor cell pool, and suggest that the UPR may represent a therapeutic target in atypical teratoid/rhabdoid tumors.


Subject(s)
Brain Neoplasms/pathology , Neural Stem Cells/cytology , Animals , Cell Lineage , Chromatin Assembly and Disassembly , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL