Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Article in English | MEDLINE | ID: mdl-21467146

ABSTRACT

DNA is folded into increasingly complex yet highly mobile structures to organize the chromosomes. In the interphase nucleus, chromosomes or part of the chromosomes encounter one another preferentially at the boundaries between chromosomal territories. Although this situation implies that the preferred chromosomal neighborhood is a key determinant of interactions between chromosomes, what this means in functional terms is currently not well understood. Using the H19 imprinting control region as a window, it has been demonstrated that epigenetic information of the primary chromatin fiber has dual functions. Thus, epigenetic marks not only influence the proximity between chromatin fibers but also transfer epigenetic states between chromatin fibers both in cis and in trans. High-throughput sequence and DNA fluorescence it situ hybridization (FISH) analyses reveal that these features require chromatin movements that are restricted in space and time. The mechanisms involved in the establishment of chromosome interactomes may provide insight of fundamental importance into pivotal regulatory processes in the nucleus, such as the coordination of transcriptional programs and replication timing.


Subject(s)
Chromosomes/metabolism , Animals , CCCTC-Binding Factor , Chromatin/metabolism , Embryoid Bodies/metabolism , Epigenesis, Genetic , Genomic Imprinting/genetics , Humans , Models, Biological , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Repressor Proteins/metabolism
2.
Curr Biol ; 11(14): 1128-30, 2001 Jul 24.
Article in English | MEDLINE | ID: mdl-11509237

ABSTRACT

The differentially methylated 5'-flank of the mouse H19 gene unidirectionally regulates the communication between enhancer elements and gene promoters and presumably represses maternal Igf2 expression in vivo [1-6]. The specific activation of the paternally inherited Igf2 allele has been proposed to involve methylation-mediated inactivation of the H19 insulator function during male germline development [1-4, 6]. Here, we addressed the role of methylation by inserting a methylated fragment of the H19-imprinting control region (ICR) into a nonmethylated episomal H19 minigene construct, followed by the transfection of ligation mixture into Hep3B cells. Individual clones were expanded and analyzed for genotype, methylation status, chromatin conformation, and insulator function. The results show that the methylated status of the H19 ICR could be propagated for several passages without spreading into the episomal vector. Moreover, the nuclease hypersensitive sites, which are typical for the maternally inherited H19 ICR allele [1], were absent on the methylated ICR, underscoring the suggestion that the methylation mark dictates parent of origin-specific chromatin conformations [1] that involve CTCF [2]. Finally, the insulator function was strongly attenuated in stably maintained episomes. Collectively, these results provide the first experimental support that the H19 insulator function is regulated by CpG methylation.


Subject(s)
CpG Islands , DNA Methylation , Genomic Imprinting , RNA, Untranslated/genetics , Alleles , Animals , Cell Line , Female , Male , Mice , Plasmids/genetics , RNA, Long Noncoding
3.
Trends Genet ; 17(9): 520-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11525835

ABSTRACT

CTCF is an evolutionarily conserved zinc finger (ZF) phosphoprotein that binds through combinatorial use of its 11 ZFs to approximately 50 bp target sites that have remarkable sequence variation. Formation of different CTCF-DNA complexes, some of which are methylation-sensitive, results in distinct functions, including gene activation, repression, silencing and chromatin insulation. Disrupting the spectrum of target specificities by ZF mutations or by abnormal selective methylation of targets is associated with cancer. CTCF emerges, therefore, as a central player in networks linking expression domains with epigenetics and cell growth regulation.


Subject(s)
DNA-Binding Proteins/physiology , Repressor Proteins , Transcription Factors/physiology , Zinc Fingers/genetics , Animals , CCCTC-Binding Factor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Environment , Gene Expression Regulation , Genes, myc , Genetics, Medical , Humans , Neoplasms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
4.
Trends Genet ; 17(3): 136-41, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11226606

ABSTRACT

Both X chromosome inactivation and autosomal genomic imprinting generate a functional hemizygosity. Here we consider models that explain the evolution of genomic imprinting and X chromosome inactivation from novel perspectives. Specifically, we suggest that random (in)activation events are common in genes and gene clusters with a low probability of transcription. These generate variability that natural selection has acted on to evolve stable monoallelic expression. Possible selection forces might include a need for dosage compensation and the prevention of biallelic silencing where a total switch off would be lethal. Two different mechanisms can accomplish regular monoallelic expression - genomic imprinting and gene counting.


Subject(s)
Dosage Compensation, Genetic , Genomic Imprinting , Animals , Biological Evolution
5.
Methods Mol Biol ; 181: 153-67, 2001.
Article in English | MEDLINE | ID: mdl-12843448

ABSTRACT

An unexpected outcome of the diploid genome is that evolutionary strategies have evolved to express only one of the alleles (1). The rapidly expanding list of genes that are expressed monoallelically fall into three main categories: random inactivation, allelic exclusion, and genomic imprinting. These categories are distinguished by whether the expressed and inactivated alleles are maintained from one cell division to the next (random inactivation occurs with each cell division, compared to stable propagation through subsequent cell divisions as seen for allelic exclusion and genomic imprinting) and whether allele inactivation or expression is determined by parent of origin of inheritance (this differentiates allelic exclusion from genomic imprinting). Genomically imprinted genes are currently more numerous than members of the other categories. This fact is likely to reflect that the persistent (in)activation of one allele in a parent of origin-specific manner has facilitated the detection of monoallelic expression patterns in RNA extracted from homogenized tissue. This crude approach does not take into account, however, different imprinted states within a tissue, or, indeed, random allelic (in)activation. It is not surprising, therefore, that well-known genes, such as Il2 (2), were only recently found to be monoallelically expressed in a random manner (1).


Subject(s)
Alleles , In Situ Hybridization/methods , Animals , Humans
6.
Int J Dev Biol ; 44(7): 785-90, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11128572

ABSTRACT

Gametic marks are stably propagated in order to manifest parent of origin-specific expression patterns of imprinted genes in the developing conceptus. Although the character of the imprint has not yet been fully elucidated, there is compelling evidence that it involves a methylation mark. This is exemplified by a region upstream of the H19 gene, which is not only methylated in a parent of origin-specific manner, but also regulates the silencing of the maternal Igf2 and paternal H19 alleles, respectively. We show here that the parental-specific methylation patterns within the differentially methylated domain (DMD) are perturbed in the soma during in vitro organogenesis. Under these conditions, the paternal DMD allele becomes partially demethylated, whereas the maternal DMD allele gains methylation. Despite these effects, there were no changes in allelic Igf2 or H19 expression patterns in the embryo. Finally, we show that although TSA derepresses the paternal H19 allele in ectoplacental cone when in vitro developed, there is no discernible effect on the methylation status of the paternally inherited 5'-flank in comparison to control samples. Collectively, this data demonstrates that the parental mark is sensitive to a subset of environmental cues and that a certain degree of plasticity of the gametic mark is tolerated without affecting the manifestation of the imprinted state.


Subject(s)
DNA Methylation , Embryo, Mammalian/metabolism , Genomic Imprinting , RNA, Untranslated/genetics , Alleles , Animals , Blotting, Southern , CpG Islands , Crosses, Genetic , DNA, Complementary/metabolism , Enzyme Inhibitors/pharmacology , Fathers , Gene Silencing , Genotype , Hydroxamic Acids/pharmacology , Mice , Mothers , Polymorphism, Single Nucleotide , RNA, Long Noncoding , RNA, Untranslated/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/metabolism , Time Factors
7.
Curr Biol ; 10(14): 853-6, 2000 Jul 13.
Article in English | MEDLINE | ID: mdl-10899010

ABSTRACT

In mammals, a subset of genes inherit gametic marks that establish parent of origin-dependent expression patterns in the soma ([1] and references therein). The currently most extensively studied examples of this phenomenon, termed genomic imprinting, are the physically linked Igf2 (insulin-like growth factor II) and H19 genes, which are expressed mono-allelically from opposite parental alleles [1] [2]. The repressed status of the maternal Igf2 allele is due to cis elements that prevent the H19 enhancers [3] from accessing the Igf2 promoters on the maternal chromosome [4] [5]. A differentially methylated domain (DMD) in the 5' flank of H19 is maintained paternally methylated and maternally unmethylated [6] [7]. We show here by gel-shift and chromatin immunopurification analyses that binding of the highly conserved multivalent factor CTCF ([8] [9] and references therein) to the H19 DMD is methylation-sensitive and parent of origin-dependent. Selectively mutating CTCF-contacting nucleotides, which were identified by methylation interference within the extended binding sites initially revealed by nuclease footprinting, abrogated the H19 DMD enhancer-blocking property. These observations suggest that molecular mechanisms of genomic imprinting may use an unusual ability of CTCF to interact with a diverse spectrum of variant target sites, some of which include CpGs that are responsible for methylation-sensitive CTCF binding in vitro and in vivo.


Subject(s)
DNA-Binding Proteins/metabolism , Muscle Proteins/genetics , RNA, Untranslated , Repressor Proteins , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , CCCTC-Binding Factor , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , Enhancer Elements, Genetic , Female , Insulin-Like Growth Factor II/genetics , Male , Mice , Molecular Sequence Data , Protein Binding , RNA, Long Noncoding , Zinc Fingers
8.
Curr Biol ; 10(8): 449-57, 2000 Apr 20.
Article in English | MEDLINE | ID: mdl-10801414

ABSTRACT

BACKGROUND: During mouse prenatal development, the neighbouring insulin-like growth factor II (Igf2) and H19 loci are expressed monoallelically from the paternal and maternal alleles, respectively. Identical spatiotemporal expression patterns and enhancer deletion experiments show that the Igf2 and H19 genes share a common set of enhancers. Deletion of a differentially methylated region in the 5' flank of the H19 gene partially relieves the repression of the maternal Igf2 and paternal H19 alleles in the soma. The mechanisms underlying the function of the 5' flank of the H19 gene are, however, unknown. RESULTS: Chromatin analysis showed that the 5' flank of the mouse H19 gene contains maternal-specific, multiple nuclease hypersensitive sites that map to linker regions between positioned nucleosomes. These features could be recapitulated in an episomal-based H19 minigene, which was propagated in human somatic cells. Although the 5' flank of the H19 promoter has no intrinsic silencer activity under these conditions, it unidirectionally extinguished promoter-enhancer communications in a position-dependent manner, without directly affecting the enhancer function. CONCLUSIONS: The unmethylated 5' flank of the H19 gene adopts an unusual and maternal-specific chromatin conformation in somatic cells and regulates enhancer-promoter communications, thereby providing an explanation for its role in manifesting the repressed state of the maternally inherited Igf2 allele.


Subject(s)
Chromatin/chemistry , Enhancer Elements, Genetic , Insulin-Like Growth Factor II/genetics , Muscle Proteins/genetics , Promoter Regions, Genetic , RNA, Untranslated , Alleles , Animals , Blotting, Southern , Cell Line , Female , Fetus , Humans , Male , Mice , Muscle Proteins/metabolism , Plasmids , Polymerase Chain Reaction , Protein Conformation , RNA, Long Noncoding , Terminal Repeat Sequences , Transformation, Genetic
9.
Mamm Genome ; 10(12): 1150-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10594239

ABSTRACT

We previously described a gene, Ipl (Tssc3), that is expressed selectively from the maternal allele in placenta, yolk sac, and fetal liver and that maps within the imprinted domain of mouse distal Chromosome (Chr) 7/human Chr 11p15.5 (Hum Mol Genet 6, 2021, 1997). Ipl is similar to TDAG51, a gene that is involved in FAS/CD95 expression. Here we describe another gene, Tih1 (TDAG/Ipl homologue 1), with equivalent sequence similarity to Ipl. Structural prediction indicates that the products of these three genes share a central motif resembling a pleckstrin-homology (PH) domain, and TIH1 protein has weak sequence similarity to the PH-domain protein SEC7/CYTOHESIN. Like Ipl, Tih1 is a small gene with a single small intron. Tih1 maps to distal mouse Chr 1 and human Chr 1q31, chromosomal regions that have not shown evidence for imprinting and, in contrast to Ipl, Tih1 is expressed equally from both parental alleles. Ipl, Tih1, and TDAG51 have overlapping but distinct patterns of expression. Tih1 and TDAG51 are expressed in multiple fetal and adult tissues. In contrast, during early mouse development Ipl mRNA and protein are highly specific for two tissues involved in maternal/fetal exchange: visceral endoderm of the yolk sac and labyrinthine trophoblast of the placenta. These findings highlight the dominance of chromosomal context over gene structure in some examples of parental imprinting and extend previous evidence for placenta-specific expression of imprinted genes. The data also define a new subfamily of PH domain genes.


Subject(s)
Blood Proteins/chemistry , Genomic Imprinting/genetics , Multigene Family/genetics , Nuclear Proteins , Phosphoproteins , Physical Chromosome Mapping , Proteins/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Blood Proteins/genetics , Chromosomes, Human, Pair 1/genetics , Codon, Initiator/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Humans , Introns/genetics , Mice , Molecular Sequence Data , Organ Specificity , Protein Conformation , Proteins/chemistry , Proteins/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcription Factors/genetics
10.
Cancer Res ; 59(16): 3889-92, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10463576

ABSTRACT

Numerous observations link the loss of imprinting of insulin-like growth factor 2 (IGF2) and an overdosage of this growth factor gene with cancer, in general, and with Wilms' tumorigenesis, in particular. It is not known, however, if loss of imprinting correlates with specific stages of neoplasia or if allelic expression patterns vary within the tumor. By applying an allele-specific in situ hybridization technique to formalin-fixed thin sections, we show that the parental IGF2 alleles can be differentially expressed, not only in Wilms' tumors, but also in nephrogenic rests (which represent premalignant lesions) of Wilms' tumor patients. Moreover, a subpopulation of mesenchymal cells, which surrounds tumor nodules, expresses IGF2 biallelically irrespective of the imprinted state of IGF2 within the tumor. These data show that Wilms' tumorigenesis involves epigenetic heterogeneity as visualized by variable allelic IGF2 expression patterns.


Subject(s)
Alleles , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/genetics , Kidney Neoplasms/genetics , Wilms Tumor/genetics , Genetic Variation , Humans
11.
Dev Biol ; 212(1): 124-36, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10419690

ABSTRACT

PDGFB is a growth factor which is vital for the completion of normal prenatal development. In this study, we report the phenotypic analysis of placentas from mouse conceptuses that lack a functional PDGFB or PDGFRbeta gene. Placentas of both types of mutant exhibit changes in the labyrinthine layer, including dilated embryonic blood vessels and reduced numbers of both pericytes and trophoblasts. These changes are seen from embryonic day (E) 13.5, which coincides with the upregulation of PDGFB mRNA levels in normal placentas. By E17, modifications in shape, size, and number of the fetal blood vessels in the mutant placentas cause an abnormal ratio of the surface areas between the fetal and the maternal blood vessels in the labyrinthine layer. Our data suggest that PDGFB acts locally to contribute to the development of the labyrinthine layer of the fetal placenta and the formation of a proper nutrient-waste exchange system during fetal development. We point out that the roles of PDGFB/Rbeta signaling in the placenta may be analogous to those in the developing kidney, by controlling pericytes in the labyrinthine layer and mesangial cells in the kidney.


Subject(s)
Placenta/anatomy & histology , Platelet-Derived Growth Factor/physiology , Proto-Oncogene Proteins/physiology , Animals , Capillaries/anatomy & histology , Female , Kidney/embryology , Maternal-Fetal Exchange , Mice , Mice, Knockout , Models, Biological , Pericytes/metabolism , Placenta/blood supply , Placenta/ultrastructure , Platelet-Derived Growth Factor/analysis , Pregnancy , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-sis , Receptors, Platelet-Derived Growth Factor/analysis , Receptors, Platelet-Derived Growth Factor/blood , Receptors, Platelet-Derived Growth Factor/physiology , Time Factors , Trophoblasts/metabolism
12.
Mech Dev ; 82(1-2): 195-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10354485

ABSTRACT

Little is known of how the genetic background effects the phenomenon of genomic imprinting. The H19 gene belongs to a cluster of imprinted genes on human chromosome 11. Here we show that the alternative splicing of a human H19 transcript is genotype-specific. Moreover, this variant transcript, which lacks exon 4, is either not found at all, is widely expressed or is confined to extra-villous cytotrophoblasts in first trimester placenta, depending on a combination of the genotype and the sex of the transmitting parent.


Subject(s)
Genes, Tumor Suppressor , Genomic Imprinting , Muscle Proteins/genetics , RNA, Untranslated , Alleles , Alternative Splicing , Base Sequence , DNA/genetics , Female , Gene Expression Regulation, Developmental , Genotype , Humans , In Situ Hybridization , Male , Oligonucleotide Probes/genetics , Pregnancy , RNA/genetics , RNA/metabolism , RNA, Long Noncoding , Trophoblasts/metabolism
14.
Dev Genes Evol ; 209(2): 113-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10022954

ABSTRACT

The IGF2 and H19 genes are genomically imprinted and expressed preferentially from the paternal and maternal alleles, respectively, during human prenatal development. The exact role of the parental imprint(s), however, is not known. To explore this issue in some detail, we have examined human androgenetic cells which by definition should be incapable of allelic discrimination given the paternal origin of both genomes. Allele-specific in situ hybridisation analysis of dispermic complete hydatidiform moles shows that IGF2 and H19 can be found to be transcriptionally active in a variegated manner, which results in the generation of random monoallelic expression patterns. This data shows that imprinted genes can be expressed monoallelically in the absence of discriminating parental marks and raises the question whether or not mechanisms underlying monoallelic expression preceded the acquisition of parental imprints during evolution.


Subject(s)
Genome, Human , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Muscle Proteins/genetics , RNA, Untranslated , Alleles , Genes, Tumor Suppressor , Humans , RNA, Long Noncoding
16.
Nat Med ; 4(11): 1276-80, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9809551

ABSTRACT

Loss of imprinting (LOI) is an epigenetic alteration of some cancers involving loss of parental origin-specific expression of imprinted genes. We observed LOI of the insulin-like growth factor-II gene in twelve of twenty-seven informative colorectal cancer patients (44%), as well as in the matched normal colonic mucosa of the patients with LOI in their cancers, and in peripheral blood samples of four patients. Ten of eleven cancers (91%) with microsatellite instability showed LOI, compared with only two of sixteen tumors (12%) without microsatellite instability (P < 0.001). Control patients without cancer showed LOI in colonic mucosa of only two of sixteen cases (12%, P < 0.001) and two of fifteen blood samples (13%, P < 0.001). These data suggest that LOI in tumor and normal tissue identifies most colorectal cancer patients with microsatellite instability in their tumors, and that LO! may identify an important subset of the population with cancer or at risk of developing cancer.


Subject(s)
Colorectal Neoplasms/genetics , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Intestinal Mucosa/metabolism , Microsatellite Repeats , Promoter Regions, Genetic , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Genetic Markers , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Polymerase Chain Reaction , Reference Values
17.
Proc Natl Acad Sci U S A ; 95(21): 12208-13, 1998 Oct 13.
Article in English | MEDLINE | ID: mdl-9770465

ABSTRACT

Nuclear receptors regulate metabolic pathways in response to changes in the environment by appropriate alterations in gene expression of key metabolic enzymes. Here, a computational search approach based on iteratively built hidden Markov models of nuclear receptors was used to identify a human nuclear receptor, termed hPAR, that is expressed in liver and intestines. hPAR was found to be efficiently activated by pregnanes and by clinically used drugs including rifampicin, an antibiotic known to selectively induce human but not murine CYP3A expression. The CYP3A drug-metabolizing enzymes are expressed in gut and liver in response to environmental chemicals and clinically used drugs. Interestingly, hPAR is not activated by pregnenolone 16alpha-carbonitrile, which is a potent inducer of murine CYP3A genes and an activator of the mouse receptor PXR.1. Furthermore, hPAR was found to bind to and trans-activate through a conserved regulatory sequence present in human but not murine CYP3A genes. These results provide evidence that hPAR and PXR.1 may represent orthologous genes from different species that have evolved to regulate overlapping target genes in response to pharmacologically distinct CYP3A activators, and have potential implications for the in vitro identification of drug interactions important to humans.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Oxidoreductases, N-Demethylating/biosynthesis , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid , Signal Transduction , Amino Acid Sequence , Base Sequence , Caco-2 Cells , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , DNA Primers , DNA, Complementary , Enzyme Induction , Humans , Molecular Sequence Data , Oxidoreductases, N-Demethylating/metabolism , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/genetics , Regulatory Sequences, Nucleic Acid , Transcriptional Activation
18.
J Biol Chem ; 273(43): 28247-52, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9774446

ABSTRACT

The imprinted H19 gene produces a fully processed transcript that does not exhibit any conserved open reading frame between mouse and man. Although transcriptional control elements associated with the mouse H19 locus have been shown to control the neighboring Igf2 gene in cis, the prevailing view is that the cytoplasmic H19 transcript does not display any function. In contrast to earlier reports, we show here that the H19 transcript is associated with polysomes in a variety of cell types, in both mouse and man. A possible trans-function of the H19 gene is suggested by a reciprocal correlation in trans between cytoplasmic H19 and IGF2 mRNA levels, as well as IGF2 mRNA translatability. We discuss these results in terms of their challenge to the prevailing dogma on the function of the enigmatic H19 gene, as well as with respect to the ontogeny of the Beckwith-Wiedemann syndrome, and propose that the human H19 gene is an antagonist of IGF2 expressivity in trans.


Subject(s)
Genes, Regulator , Insulin-Like Growth Factor II/biosynthesis , Polyribosomes/metabolism , RNA, Messenger/genetics , Animals , Beckwith-Wiedemann Syndrome/etiology , Gene Expression Regulation , Genomic Imprinting , Humans , Insulin-Like Growth Factor II/genetics , Mice , Models, Genetic , Polyribosomes/genetics , Protein Biosynthesis , RNA, Messenger/metabolism , Wilms Tumor/genetics
19.
Oncogene ; 16(1): 113-9, 1998 Jan 08.
Article in English | MEDLINE | ID: mdl-9467950

ABSTRACT

The IGF2 gene, which encodes a growth factor, is subject to genomic imprinting. The frequently observed loss of IGF2 imprinting in a variety of tumors has been suggested to contribute to neoplasia. Since these reports have not documented the imprinting status of IGF2 at the cellular level, it cannot be excluded that the imprinting status might vary within the tumor. The possibility that loss of IGF2 imprinting in neoplastic cells reflects random imprinting patterns, was therefore addressed. We show here that individual cell populations of the JEG-3 choriocarcinoma cell line display heterogenous imprinting patterns of both IGF2 and H19. In addition, a lack of correlation between IGF2 and H19 imprinting status suggests that any regional parental imprint has been functionally lost. This notion is reinforced by the observation that JEG-3 cell subclones display a range of promoter-specific IGF2 allele usage. Moreover, we observed that the imprinting status of H19 and IGF2 were differentially modulated in JEG-3-derived tumors generated in nude mice. The results suggest that allele-specific expression of IGF2 operates in the absence of a parental imprint. Finally, our observations urge caution with respect to the general interpretation of biallelic expression as 'loss of imprinting'.


Subject(s)
Alleles , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , RNA, Untranslated , Animals , Choriocarcinoma/genetics , Choriocarcinoma/pathology , Mice , Mice, Nude , Muscle Proteins/genetics , Promoter Regions, Genetic , RNA, Long Noncoding , Tumor Cells, Cultured
20.
Development ; 125(1): 61-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9389664

ABSTRACT

Transcriptional silencing can reflect heritable, epigenetic inactivation of genes, either singly or in groups, during the life-time of an organism. This phenomenon is exemplified by parent-of-origin-specific inactivation events (genomic imprinting) for a subset of mammalian autosomal genes, such as H19. Very little is known, however, about the timing and mechanism(s) of silencing of the paternal H19 allele during mouse development. Using a novel in situ approach, we present evidence that the silencing of the paternal H19 allele is progressive in the trophectodermal lineage during early mouse development and generates variegated expression patterns. The silencing process apparently involves recruitment of histone deacetylases since the mosaic paternal-specific H19 expression reappears in trichostatin A-treated mouse conceptuses, undergoing in vitro organogenesis. Moreover, the paternal H19 alleles of PatDup.d7 placentas, in which a region encompassing the H19 locus of chromosome 7 is bipaternally derived, partially escape the silencing process and are expressed in a variegated manner. We suggest that allele-specific silencing of H19 share some common features with chromatin-mediated silencing in position-effect variegation.


Subject(s)
Gene Expression Regulation, Developmental , Genomic Imprinting , Histones/metabolism , Muscle Proteins/genetics , RNA, Untranslated , Acetylation , Alleles , Animals , Embryonic and Fetal Development/drug effects , Embryonic and Fetal Development/physiology , Enzyme Inhibitors/pharmacology , Genotype , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , In Situ Hybridization , Mice , Phenotype , Placenta/cytology , RNA/analysis , RNA, Long Noncoding , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...