Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 94(4): 95, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26984997

ABSTRACT

The nucleolus is a dynamic nuclear compartment that is mostly involved in ribosome subunit biogenesis; however, it may also play a role in many other biological processes, such as stress response and the cell cycle. Mainly using electron microscopy, several studies have tried to decipher how active nucleoli are set up during early development in mice. In this study, we analyzed nucleologenesis during mouse early embryonic development using 3D-immunofluorescent detection of UBF and Nopp140, two proteins associated with different nucleolar compartments. UBF is a transcription factor that helps maintain the euchromatic state of ribosomal genes; Nopp140 is a phosphoprotein that has been implicated in pre-rRNA processing. First, using detailed image analyses and the in situ proximity ligation assay technique, we demonstrate that UBF and Nopp140 dynamic redistribution between the two-cell and blastocyst stages (time of implantation) is correlated with morphological and structural modifications that occur in embryonic nucleolar compartments. Our results also support the hypothesis that nucleoli develop at the periphery of nucleolar precursor bodies. Finally, we show that the RNA polymerase I inhibitor CX-5461: 1) disrupts transcriptional activity, 2) alters preimplantation development, and 3) leads to a complete reorganization of UBF and Nopp140 distribution. Altogether, our results underscore that highly dynamic changes are occurring in the nucleoli of embryos and confirm a close link between ribosomal gene transcription and nucleologenesis during the early stages of development.


Subject(s)
DNA, Ribosomal/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Animals , Benzothiazoles , Female , Mice, Inbred C57BL , Naphthyridines , RNA Polymerase I/antagonists & inhibitors
2.
BMC Dev Biol ; 12: 30, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-23095683

ABSTRACT

BACKGROUND: Embryonic development proceeds through finely tuned reprogramming of the parental genomes to form a totipotent embryo. Cells within this embryo will then differentiate and give rise to all the tissues of a new individual. Early embryonic development thus offers a particularly interesting system in which to analyze functional nuclear organization. When the organization of higher-order chromatin structures, such as pericentromeric heterochromatin, was first analyzed in mouse embryos, specific nuclear rearrangements were observed that correlated with embryonic genome activation at the 2-cell stage. However, most existing analyses have been conducted by visual observation of fluorescent images, in two dimensions or on z-stack sections/projections, but only rarely in three dimensions (3D). RESULTS: In the present study, we used DNA fluorescent in situ hybridization (FISH) to localize centromeric (minor satellites), pericentromeric (major satellites), and telomeric genomic sequences throughout the preimplantation period in naturally fertilized mouse embryos (from the 1-cell to blastocyst stage). Their distribution was then analyzed in 3D on confocal image stacks, focusing on the nucleolar precursor bodies and nucleoli known to evolve rapidly throughout the first developmental stages. We used computational imaging to quantify various nuclear parameters in the 3D-FISH images, to analyze the organization of compartments of interest, and to measure physical distances between these compartments. CONCLUSIONS: The results highlight differences in nuclear organization between the two parental inherited genomes at the 1-cell stage, i.e. just after fertilization. We also found that the reprogramming of the embryonic genome, which starts at the 2-cell stage, undergoes other remarkable changes during preimplantation development, particularly at the 4-cell stage.


Subject(s)
Cell Nucleus/metabolism , Embryo, Mammalian/cytology , Embryonic Development , Zygote/cytology , Animals , Cell Nucleolus/metabolism , Cell Nucleus/physiology , Cell Nucleus Shape , Cell Polarity , Centromere/genetics , Centromere/metabolism , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Female , Heterochromatin/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Telomere/genetics , Telomere/metabolism
3.
PLoS One ; 7(5): e38156, 2012.
Article in English | MEDLINE | ID: mdl-22693592

ABSTRACT

BACKGROUND: In the mouse zygote, DNA methylation patterns are heavily modified, and differ between the maternal and paternal pronucleus. Demethylation of the paternal genome has been described as an active and replication-independent process, although the mechanisms responsible for it remain elusive. Recently, 5-hydroxymethylcytosine has been suggested as an intermediate in this demethylation. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we quantified DNA methylation and hydroxymethylation in both pronuclei of the mouse zygote during the replication period and we examined their patterns on the pericentric heterochromatin using 3D immuno-FISH. Our results demonstrate that 5-methylcytosine and 5-hydroxymethylcytosine localizations on the pericentric sequences are not complementary; indeed we observe no enrichment of either marks on some regions and an enrichment of both on others. In addition, we show that DNA demethylation continues during DNA replication, and is inhibited by aphidicolin. Finally, we observe notable differences in the kinetics of demethylation and hydroxymethylation; in particular, a peak of 5-hydroxymethylcytosine, unrelated to any change in 5-methylcytosine level, is observed after completion of replication. CONCLUSIONS/SIGNIFICANCE: Together our results support the already proposed hypothesis that 5-hydroxymethylcytosine is not a simple intermediate in an active demethylation process and could play a role of its own during early development.


Subject(s)
5-Methylcytosine/metabolism , Cytosine/analogs & derivatives , Zygote/metabolism , Animals , Cell Cycle , Cytosine/metabolism , DNA Methylation , DNA Replication , Female , Heterochromatin/metabolism , Male , Mice , Time Factors , Zygote/cytology
4.
J Reprod Dev ; 58(4): 467-75, 2012.
Article in English | MEDLINE | ID: mdl-22572731

ABSTRACT

Phosphorylation of histone H3 at Ser10 (H3S10P) has been linked to a variety of cellular processes, such as chromosome condensation and gene activation/silencing. Remarkably, in mammalian somatic cells, H3S10P initiates in the pericentromeric heterochromatin during the late G2 phase, and phosphorylation spreads throughout the chromosomes arms in prophase, being maintained until the onset of anaphase when it gets dephosphorylated. Considerable studies have been carried out about H3S10P in different organisms; however, there is little information about this histone modification in mammalian embryos. We hypothesized that this epigenetic modification could also be a marker of pericentromeric heterochromatin in preimplantation embryos. We therefore followed the H3S10P distribution pattern in the G1/S and G2 phases through the entire preimplantation development in in vivo mouse embryos. We paid special attention to its localization relative to another pericentromeric heterochromatin marker, HP1ß and performed immunoFISH using specific pericentromeric heterochromatin probes. Our results indicate that H3S10P presents a remarkable distribution pattern in preimplantation mouse embryos until the 4-cell stage and is a better marker of pericentromeric heterochromatin than HP1ß. After the 8-cell stage, H3S10P kinetic is more similar to the somatic one, initiating during G2 in chromocenters and disappearing upon telophase. Based on these findings, we believe that H3S10P is a good marker of pericentromeric heterochromatin, especially in the late 1- and 2-cell stages as it labels both parental genomes and that it can be used to further investigate epigenetic regulation and heterochromatin mechanisms in early preimplantation embryos.


Subject(s)
Blastocyst/metabolism , Embryonic Development , Epigenesis, Genetic , Heterochromatin/metabolism , Histones/metabolism , Interphase , Serine/metabolism , Animals , Biomarkers/metabolism , Blastocyst/cytology , Female , In Situ Hybridization, Fluorescence , Metaphase , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Phosphorylation , Pregnancy , Prophase , Protein Processing, Post-Translational , Telophase
5.
Int J Dev Biol ; 56(10-12): 877-87, 2012.
Article in English | MEDLINE | ID: mdl-23417410

ABSTRACT

During the final step of oogenesis, the oocyte nucleus is subject to large-scale modifications that correlate with transcriptional silencing. While oocytes with dense chromatin around the nucleolus are silent (SN, surrounded nucleolus), oocytes with uncondensed chromatin (NSN, non-surrounded nucleolus) are transcriptionally active. It is believed that epigenetic mechanisms that participate in gene expression regulation could play a role in this event. In this context, we examined the behaviour of heterochromatin and related histone modifications during the NSN to SN transition by immunostaining. Using fluorescent in situ hybridization on three dimensional-preserved nuclei (3D-FISH), we also studied the distribution of centromeric, pericentromeric and ribosomal (rDNA) sequences in relation to the nucleolus (also called the nucleolus-like body, NLB). We observed that in NSN-type oocytes, pericentromeric heterochromatin is aggregated within chromocenters. In SN-type oocytes, pericentromeric heterochromatin and centromeres form a discontinuous ring around the NLB. rDNA sequences, which initially present a pearl necklace structure, gather together in seven highly condensed foci at the NLB periphery. H3K9me3 and H4K20me3 heterochromatin marks clearly label chromocenters, whereas H3K4me3 and H4K5ac are totally excluded from heterochromatin regions, even in the very compact SN-nuclei. Remarkably, H3K27me3 displays an intermediate behavior. It appears that GV oocyte nuclei exhibit a specific epigenetic landscape. Histone modifications, related to both active and repressive chromatin structures, seem to follow the large-scale chromatin movements that occur during the NSN to SN transition. We also demonstrate that, while heterochromatin regions re-localize around the NLB, rDNA sequences adopt a highly compact structure in SN-type oocytes.


Subject(s)
Cell Nucleus/genetics , Epigenesis, Genetic , Genome/genetics , Oocytes/metabolism , Animals , Cell Nucleolus/genetics , Centromere/genetics , Chromatin/genetics , Chromatin/metabolism , DNA, Ribosomal/genetics , Epigenomics , Female , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/metabolism , In Situ Hybridization, Fluorescence , Methylation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Microscopy, Fluorescence , Oocytes/cytology , Oogenesis/genetics
6.
Biol Aujourdhui ; 204(3): 205-13, 2010.
Article in French | MEDLINE | ID: mdl-20950564

ABSTRACT

In mammals, the embryonic genome is first transcriptionally inactive after fertilization. Embryonic development is then strictly dependent on the maternally inherited RNA and proteins accumulated before ovulation and present in the oocyte cytoplasm. The onset of embryonic gene expression is initiated later during development, i.e. during the "embryonic genome activation (EGA)". EGA takes place at various preimplantation stages according to species and is dependent on the presence of the basal transcriptional machinery components but also on parental genomes reorganizations after fertilization. Indeed, during the first embryonic cycles, nuclei undergo intense remodeling that could be a key regulator of embryonic development.


Subject(s)
Embryo, Mammalian/physiology , Fertilization/physiology , Mammals/genetics , Mammals/physiology , Animals , Blastocyst/physiology , Cell Nucleus/genetics , Cell Nucleus/physiology , DNA Modification Methylases/genetics , Female , Fertilization/genetics , Genetic Variation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Pregnancy , Transcription, Genetic , Transcriptional Activation
7.
Methods Mol Biol ; 659: 401-8, 2010.
Article in English | MEDLINE | ID: mdl-20809330

ABSTRACT

A common problem in research laboratories that study the mammalian embryo is the limited supply of live material. For this reason, new methods are constantly being developed and existing methods for in vitro models using cells in culture are being adapted to represent embryogenesis. Three-dimensional fluorescence in situ hybridization (3D-FISH) is an important tool to study where genomic sequences are positioned within nuclei without interfering with this 3D organization. When used in the embryo, this technique provides vital information about the distribution of specific sequences in relation to embryonic nuclear substructures such as nucleolar precursor bodies and chromocenters. In this chapter, we will present a detailed description of FISH in order to perform 3D-FISH in the early preimplantation murine embryos.


Subject(s)
DNA Probes/genetics , DNA Probes/metabolism , Embryo, Mammalian/metabolism , In Situ Hybridization, Fluorescence/methods , Repetitive Sequences, Nucleic Acid , Animals , Color , Mice , Tissue Embedding
8.
PLoS Comput Biol ; 6(7): e1000853, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20628576

ABSTRACT

In eukaryotes, the interphase nucleus is organized in morphologically and/or functionally distinct nuclear "compartments". Numerous studies highlight functional relationships between the spatial organization of the nucleus and gene regulation. This raises the question of whether nuclear organization principles exist and, if so, whether they are identical in the animal and plant kingdoms. We addressed this issue through the investigation of the three-dimensional distribution of the centromeres and chromocenters. We investigated five very diverse populations of interphase nuclei at different differentiation stages in their physiological environment, belonging to rabbit embryos at the 8-cell and blastocyst stages, differentiated rabbit mammary epithelial cells during lactation, and differentiated cells of Arabidopsis thaliana plantlets. We developed new tools based on the processing of confocal images and a new statistical approach based on G- and F- distance functions used in spatial statistics. Our original computational scheme takes into account both size and shape variability by comparing, for each nucleus, the observed distribution against a reference distribution estimated by Monte-Carlo sampling over the same nucleus. This implicit normalization allowed similar data processing and extraction of rules in the five differentiated nuclei populations of the three studied biological systems, despite differences in chromosome number, genome organization and heterochromatin content. We showed that centromeres/chromocenters form significantly more regularly spaced patterns than expected under a completely random situation, suggesting that repulsive constraints or spatial inhomogeneities underlay the spatial organization of heterochromatic compartments. The proposed technique should be useful for identifying further spatial features in a wide range of cell types.


Subject(s)
Cell Nucleus/chemistry , Centromere/chemistry , Heterochromatin/chemistry , Imaging, Three-Dimensional , Models, Statistical , Animals , Arabidopsis/cytology , Embryo, Mammalian/cytology , Female , Mammary Glands, Animal/cytology , Microscopy, Confocal , Monte Carlo Method , Nuclear Proteins/chemistry , Rabbits
9.
C R Biol ; 332(11): 937-46, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19909917

ABSTRACT

Compartmentalization is one of the fundamental principles which underly nuclear function. Numerous studies describe complex and sometimes conflicting relationships between nuclear gene positioning and transcription regulation. Therefore the question is whether topological landmarks and/or organization principles exist to describe the nuclear architecture and, if existing, whether these principles are identical in the animal and plant kingdoms. In the frame of an agroBI-INRA program on nuclear architecture, we set up a multidisciplinary approach combining biological studies, spatial statistics and 3D modeling to investigate spatial organization of a nuclear compartment in both plant and animal cells in their physiological contexts. In this article, we review the questions addressed in this program and the methodology of our work.


Subject(s)
Cell Nucleus/ultrastructure , Eukaryotic Cells/ultrastructure , Models, Biological , Plant Cells , Algorithms , Animals , Arabidopsis/cytology , Blastocyst/cytology , Cell Compartmentation , Cell Differentiation , Cell Nucleus/physiology , Eukaryotic Cells/physiology , Female , Gene Expression Regulation , Gene Expression Regulation, Plant , Image Processing, Computer-Assisted , Mammary Glands, Animal/cytology , Plants/genetics , Pregnancy , Protoplasts/ultrastructure , Rabbits , Systems Biology/methods
10.
BMC Dev Biol ; 9: 11, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19210795

ABSTRACT

BACKGROUND: Genome reprogramming in early mouse embryos is associated with nuclear reorganization and particular features such as the peculiar distribution of centromeric and pericentric heterochromatin during the first developmental stage. This zygote-specific heterochromatin organization could be observed both in maternal and paternal pronuclei after natural fertilization as well as in embryonic stem (ES) cell nuclei after nuclear transfer suggesting that this particular type of nuclear organization was essential for embryonic reprogramming and subsequent development. RESULTS: Here, we show that remodeling into a zygotic-like organization also occurs after somatic cell nuclear transfer (SCNT), supporting the hypothesis that reorganization of constitutive heterochromatin occurs regardless of the source and differentiation state of the starting material. However, abnormal nuclear remodeling was frequently observed after SCNT, in association with low developmental efficiency. When transient treatment with the histone deacetylase inhibitor trichostatin A (TSA) was tested, we observed improved nuclear remodeling in 1-cell SCNT embryos that correlated with improved rates of embryonic development at subsequent stages. CONCLUSION: Together, the results suggest that proper organization of constitutive heterochromatin in early embryos is involved in the initial developmental steps and might have long term consequences, especially in cloning procedures.


Subject(s)
Chromatin Assembly and Disassembly , Embryonic Development/drug effects , Heterochromatin/metabolism , Hydroxamic Acids/pharmacology , Animals , Cell Cycle , Embryo, Mammalian/metabolism , Mice , Nuclear Transfer Techniques
11.
Mol Reprod Dev ; 73(9): 1102-11, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16736527

ABSTRACT

The ability of cloned embryos to sustain full-term development depends on the ability of the recipient ooplasm to reprogram the donor cell genome. As the nuclear architecture has recently emerged as a key-factor in the regulation of gene expression, we questioned whether early embryos obtained from transfer of ES metaphasic chromosomes into mouse ooplasm would adopt the somatic or embryonic type of nuclear organization. We have particularly focused on the arrangement of chromosomal territories with respect to the nucleolar compartment, and the pericentric heterochromatin domains called chromocenters. We found that nuclear transfer triggers profound chromatin rearrangements including the dispersion of the donor cell chromocenters components. These rearrangements lead to a typical 1-cell pronuclear organization, namely a radial arrangement of the chromosome territories with centromeres attached to the nucleoli, which adopt the compact fibrillar structure of nucleolar precursor bodies (NPBs). Subsequently, during the second cycle, the cloned embryos undergo further reorganization with the establishment of new chromocenters, clustered in one part of the nucleus, as during normal embryogenesis. We could also establish that the adequate distribution of chromosomal territories at the pronuclear stage seems important for the development until blastocyst.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/physiology , Genome/genetics , Nuclear Transfer Techniques , Oocytes/cytology , Oocytes/metabolism , Animals , Cells, Cultured , Chromatin/genetics , Chromosomes, Mammalian/genetics , Cytoplasm/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Mammalian/ultrastructure , Female , Mice , Microscopy, Electron , Mitosis , Time Factors
12.
Dev Biol ; 292(2): 317-32, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16680825

ABSTRACT

Although a growing number of studies investigates functional genome organization in somatic cell nuclei, it is largely unknown how mammalian genome organization is established during embryogenesis. To address this question, we investigated chromo center formation and the peculiar arrangements of chromosome domains in early mouse embryos. At the one-cell stage, we observed characteristic arrangements of chromosomes and chromo center components. Subsequently, starting with the burst of zygotic genome transcription major rearrangements led to the establishment of somatic type chromo centers with a defined spatio-temporal organization. These processes appeared to be completed at the blastocyst stage with the onset of cell differentiation. During the same developmental period, a fraction of pericentric heterochromatin that was late replicating in the first cycle underwent switches in replication timing, spatial organization and epigenetic marks. Cloning experiments revealed that the genome organization typical for more advanced stages was quickly reverted into the one-cell stage-specific form after nuclear transfer, supporting the idea that reprogramming associated genome remodeling in normal and cloned embryos is determined by cytoplasmic factors. Together, the results suggest that distinct but characteristic forms of nuclear genome organization are required for genome reprogramming in early embryos and for proper regulation of differential gene expression patterns at later stages.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Genome , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cattle , Cell Nucleolus/metabolism , Centromere/metabolism , Chorionic Gonadotropin/pharmacology , Chromatin/metabolism , Chromosomes/metabolism , DNA/metabolism , Female , Fertilization in Vitro , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique , Fluorescent Dyes , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Models, Biological , Nuclear Transfer Techniques , Oocytes/cytology , Pregnancy , Rhodamines , Time Factors
13.
J Biol Chem ; 281(4): 2151-61, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16301322

ABSTRACT

Amyloid deposits within the cerebral tissue constitute a characteristic lesion associated with Alzheimer disease. They mainly consist of the amyloid peptide Abeta and display an abnormal content in Zn(2+) ions, together with many truncated, isomerized, and racemized forms of Abeta. The region 1-16 of Abeta can be considered the minimal zinc-binding domain and contains two aspartates subject to protein aging. The influence of zinc binding and protein aging related modifications on the conformation of this region of Abeta is of importance given the potentiality of this domain to constitute a therapeutic target, especially for immunization approaches. In this study, we determined from NMR data the solution structure of the Abeta-(1-16)-Zn(2+) complex in aqueous solution at pH 6.5. The residues His(6), His(13), and His(14) and the Glu(11) carboxylate were identified as ligands that tetrahedrally coordinate the Zn(II) cation. In vitro aging experiments on Abeta-(1-16) led to the formation of truncated and isomerized species. The major isomer generated, Abeta-(1-16)-l-iso-Asp(7), displayed a local conformational change in the His(6)-Ser(8) region but kept a zinc binding propensity via a coordination mode involving l-iso-Asp(7). These results are discussed here with regard to Abeta fibrillogenesis and the potentiality of the region 1-16 of Abeta to be used as a therapeutic target.


Subject(s)
Aging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Zinc/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Ions/chemistry , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Temperature
14.
J Cell Biochem ; 96(3): 611-21, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16088957

ABSTRACT

The expression of casein genes is specific to the mammary gland and maximal during lactation. However, among the numerous mammary cell lines described so far, only a few express some casein genes. The regulatory regions of casein genes have been largely described but the mechanisms explaining the mammary specific expression of these genes, and their silencing in most mammary cell lines, have not yet been fully elucidated. To test the hypothesis that the nuclear location of the casein genes may affect their expression, we transfected HC11 mouse mammary cell line with a 100 kb DNA fragment surrounding the rabbit alpha S1 casein gene. We derived stable clones which express or not the transfected rabbit casein gene, in the same cellular context, independently of the number of transgene copies. Metaphase spreads were prepared from the different clones and the transfected genes were localized. Unexpectedly, we observed that in the original HC11 cell line the number of chromosomes per metaphase spread is close to 80, suggesting that HC11 cells have undergone a duplication event, since the mouse karyotype is 2n = 40. In alpha S1 casein expressing cells, the expression level does not clearly correlate with a localization of the transfected DNA proximal to the centromeres or the telomeres. Analysis of the localization of the transfected DNA in nuclear halos allows us to conclude that when expressed, transfected DNA is more closely linked to the nuclear matrix. The next step will be to study the attachment of the endogenous casein gene in mammary nuclei during lactation.


Subject(s)
Caseins/genetics , Caseins/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Mammary Glands, Animal/cytology , Nuclear Matrix/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromosomes , DNA/metabolism , Epithelial Cells/cytology , Female , Gene Dosage , In Situ Hybridization , Mice , Rabbits , Transgenes
15.
Med Sci (Paris) ; 21(4): 412-21, 2005 Apr.
Article in French | MEDLINE | ID: mdl-15811307

ABSTRACT

The fact that the nucleus of a differentiated somatic cell can be reprogrammed in order to sustain embryonic development is now well established. Experiments of somatic cell nuclear transfer (cloning) have proved that a foreign nucleus introduced into an enucleated oocyte can give rise to physiologically normal offsprings, with a normal lifespan. Such evidence of genome expression plasticity is also observed experimentally with heterokaryons, created by the fusion or the nuclear transfer between two somatic cells, where differentiated nuclei are able to express genes characteristic of the host cell. However, the epigenetic mechanisms that permit nuclear plasticity remain poorly understood. In this paper we present the main evidences showing important modifications of the large scale organisation of chromosomal domains and of the DNA methylation pattern upon nuclear transfer and during the first cleavages. These modifications of epigenetic marks, brought by an intimate contact between the chromatin and the recipient oocyte cytoplasmic factors, appear essential for further development. They are established over the first cell cycles of development. The onset of embryonic genome activation and the first cellular differentiation events that occur over the implantation period are two additional check-points of reprogramming that appear to be also highly dependent on epigenetic alterations. Beyond those stages, defective placental functions might be directly responsible for the fetal and postnatal physiopathologies frequently observed in cloned animals. No direct link between preimplantation reprogramming defaults, placental dysfunctions and low development to term has been established yet. The epigenetics studies which are now used to characterise loci specific and probably genotype dependent alterations in cloned animals of different species will provide invaluable help to define the role of epigenesis in the achievement of a developmental program.


Subject(s)
Cell Nucleus/genetics , Epigenesis, Genetic/genetics , Animals , DNA Methylation , Embryonic Development/genetics
16.
Protein Sci ; 13(12): 3151-60, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15537751

ABSTRACT

The conformational conversion of the nonpathogenic "cellular" prion isoform into a pathogenic "scrapie" protease-resistant isoform is a fundamental event in the onset of transmissible spongiform encephalopathies (TSE). During this pathogenic conversion, helix H1 and its two flanking loops of the normal prion protein are thought to undergo a conformational transition into a beta-like structure. A peptide spanning helix H1 and beta-strand S2 (residues 142-166 in human numbering) was studied by circular dichroism and nuclear magnetic resonance spectroscopies. This peptide in aqueous solution, in contrast to many prion fragments studied earlier (1) is highly soluble and (2) does not aggregate until the millimolar concentration range, and (3) exhibits an intrinsic propensity to a beta-hairpin-like conformation at neutral pH. We found that this peptide can also fold into a helix H1 conformation when dissolved in a TFE/PB mixture. The structures of the peptide calculated by MD showed solvent-dependent internal stabilizing forces of the structures and evidenced a higher mobility of the residues following the end of helix H1. These data suggest that the molecular rearrangement of this peptide in region 152-156, particularly in position 155, could be associated with the pathogenic conversion of the prion protein.


Subject(s)
Peptide Fragments/chemistry , PrPSc Proteins/chemistry , Animals , Circular Dichroism , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , PrPSc Proteins/genetics , Protein Conformation , Sheep/genetics , Solubility , Solvents/chemistry
17.
Biochem Biophys Res Commun ; 321(2): 324-8, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15358179

ABSTRACT

Amyloid plaques associated to Alzheimer's disease present a high content of zinc ions. We previously showed that the N-terminal region of the amyloid peptide Abeta constitutes an autonomous zinc-binding domain. This region encompasses the previously identified epitope Abeta(4-10) targeted by antibodies capable to reduce amyloid deposition, but the influence of Abeta/Zn binding on the epitope recognition remains unknown. We demonstrate here the effect of Zn2+ ions on the recognition of peptides sharing the sequence of the Abeta N-terminal domain, by two monoclonal antibodies recognizing the beta-amyloid(4-10) epitope. The presence of Zn2+, but not of other cations, increased the recognition of the (1-16) peptide, while it was without effect on the recognition of the (1-10) peptide. These findings show a zinc-induced conformational change of the (1-16)-N-terminal region of AP3, which results in a better accessibility of the Abeta(4-10) epitope to the anti-Abeta antibodies, and suggest a role of zinc in epitope-based vaccination approaches.


Subject(s)
Amyloid beta-Peptides/agonists , Amyloid beta-Peptides/immunology , Antibodies/immunology , Epitopes/immunology , Zinc/metabolism , Amyloid beta-Peptides/metabolism , Enzyme-Linked Immunosorbent Assay , Metals/metabolism , Metals/pharmacology , Zinc/pharmacology
18.
Proc Natl Acad Sci U S A ; 101(28): 10254-9, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15240887

ABSTRACT

Prion diseases are associated with the conversion of the alpha-helix rich prion protein (PrPC) into a beta-structure-rich insoluble conformer (PrPSc) that is thought to be infectious. The mechanism for the PrPC-->PrPSc conversion and its relationship with the pathological effects of prion diseases are poorly understood, partly because of our limited knowledge of the structure of PrPSc. In particular, the way in which mutations in the PRNP gene yield variants that confer different susceptibilities to disease needs to be clarified. We report here the 2.5-A-resolution crystal structures of three scrapie-susceptibility ovine PrP variants complexed with an antibody that binds to PrPC and to PrPSc; they identify two important features of the PrPC-->PrPSc conversion. First, the epitope of the antibody mainly consists of the last two turns of ovine PrP second alpha-helix. We show that this is a structural invariant in the PrPC-->PrPSc conversion; taken together with biochemical data, this leads to a model of the conformational change in which the two PrPC C-terminal alpha-helices are conserved in PrPSc, whereas secondary structure changes are located in the N-terminal alpha-helix. Second, comparison of the structures of scrapie-sensitivity variants defines local changes in distant parts of the protein that account for the observed differences of PrPC stability, resistant variants being destabilized compared with sensitive ones. Additive contributions of these sensitivity-modulating mutations to resistance suggest a possible causal relationship between scrapie resistance and lowered stability of the PrP protein.


Subject(s)
Epitopes/immunology , PrPC Proteins/chemistry , PrPC Proteins/immunology , PrPSc Proteins/chemistry , PrPSc Proteins/immunology , Scrapie/immunology , Animals , Antibody Specificity , Antigen-Antibody Reactions , Crystallography , Mice , Mutation , PrPC Proteins/genetics , PrPSc Proteins/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Sheep
19.
Biol Reprod ; 71(4): 1330-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15189833

ABSTRACT

We previously described that mouse oocytes and preimplantation embryos express the two subunits of interferon-gamma receptor. We now report that, despite the presence of STAT1 (signal transducer and activator of transcription 1) at both the mRNA and protein levels, interferon gamma (IFNgamma) as well as IFNalpha are unable to trigger massive nuclear translocation of STAT1 in these cells, even at high cytokine concentrations. Conversely, nuclear accumulation of STAT1 was readily observed in murine L929 somatic cells under the same conditions. However, in the absence of any stimulation, both tyrosine (Y701p) and serine (S727p) phosphorylated forms of STAT1 were already detected in the nuclei of oocytes and early embryos. Phosphorylated STAT1 appeared concentrated in large nuclear dots, which were identified by indirect immunofluorescence and electron microscopy as clusters of interchromatin granules (IGCs or speckles). A similar distribution was also observed for the serine (S727p) phosphorylated form of STAT3 as well as for tyrosine (Y689p) phosphorylated STAT2. Western blot analysis confirmed that STAT factors present in mouse oocytes are predominantly phosphorylated. In parallel, we showed that the transcription of two IFNgamma-target genes, namely interferon regulatory factor-1 (IRF-1) and suppressor of cytokine signaling-1 (SOCS-1) is indeed increased in two-cell embryos in response to IFNgamma. Altogether, our results suggest that, despite the lack of massive nuclear accumulation of STAT1 in response to exogenous IFNs and the permanent presence of phosphorylated STATs in the nucleus, JAK/ STAT pathways are functional during early development.


Subject(s)
Blastocyst/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Interferon-gamma/physiology , Oocytes/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA-Binding Proteins/genetics , Female , Interferon Regulatory Factor-1 , Interferon-alpha/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Pregnancy , Protein Transport/physiology , RNA, Messenger/analysis , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT1 Transcription Factor , Subcellular Fractions/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/genetics , Transcription, Genetic/physiology , Transcriptional Activation
20.
Hum Reprod ; 18(7): 1494-503, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832378

ABSTRACT

BACKGROUND: Using fluorescence imaging, an a posteriori multiparametric analysis was performed of human oocytes which failed to give pronucleated zygotes after IVF in cases of very low rates of fertilization or complete fertilization failure. METHODS: The analysis included: (i) the state of the maternal and paternal chromatin; (ii) quality of the metaphase II oocytes; and (iii) cortical granule (CG) distribution. RESULTS: Most oocytes were arrested in metaphase II, but they were abnormal in 50% of cases. The incidence of spindle and chromosome aberrations was strongly influenced by maternal age (69% for 40- to 45-year-old women versus 35% for 26- to 33-year-olds), and sperm chromatin was always condensed in immature oocytes, and fully decondensed only in normal metaphase II. The migration of CGs appeared to be associated with achievement of nuclear maturation at the time of puncture. CONCLUSIONS: These factors, when analysed on a complete set of oocytes from the same patient, provided information about potential causes of IVF failure, and also represented part of an 'oocyte quality evaluation' to select the assisted fertilization technique most suitable for each patient. For example, when the majority of oocytes were judged non-fertilizable at a first attempt, no pregnancy was registered at any subsequent attempt.


Subject(s)
Chromosome Aberrations , Fertilization in Vitro , Infertility, Female/pathology , Oocytes/pathology , Age Distribution , Chromatin/pathology , Female , Humans , Incidence , Infertility, Female/epidemiology , Infertility, Female/genetics , Male , Metaphase , Microtubules , Pregnancy , Sperm-Ovum Interactions , Spermatozoa , Treatment Failure
SELECTION OF CITATIONS
SEARCH DETAIL
...