Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 19(7): e1011479, 2023 07.
Article in English | MEDLINE | ID: mdl-37428812

ABSTRACT

Buruli ulcer is a chronic infectious disease caused by Mycobacterium ulcerans. The pathogen persistence in host skin is associated with the development of ulcerative and necrotic lesions leading to permanent disabilities in most patients. However, few of diagnosed cases are thought to resolve through an unknown self-healing process. Using in vitro and in vivo mouse models and M. ulcerans purified vesicles and mycolactone, we showed that the development of an innate immune tolerance was only specific to macrophages from mice able to heal spontaneously. This tolerance mechanism depends on a type I interferon response and can be induced by interferon beta. A type I interferon signature was further detected during in vivo infection in mice as well as in skin samples from patients under antibiotics regiment. Our results indicate that type I interferon-related genes expressed in macrophages may promote tolerance and healing during infection with skin damaging pathogen.


Subject(s)
Buruli Ulcer , Interferon Type I , Mycobacterium ulcerans , Mice , Animals , Buruli Ulcer/microbiology , Macrophages , Macrolides , Immune Tolerance
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074915

ABSTRACT

An important characteristic of cell differentiation is its stability. Only rarely do cells or their stem cell progenitors change their differentiation pathway. If they do, it is often accompanied by a malfunction such as cancer. A mechanistic understanding of the stability of differentiated states would allow better prospects of alleviating the malfunctioning. However, such complete information is yet elusive. Earlier experiments performed in Xenopus oocytes to address this question suggest that a cell may maintain its gene expression by prolonged binding of cell type-specific transcription factors. Here, using DNA competition experiments, we show that the stability of gene expression in a nondividing cell could be caused by the local entrapment of part of the general transcription machinery in transcriptionally active regions. Strikingly, we found that transcriptionally active and silent forms of the same DNA template can stably coexist within the same nucleus. Both DNA templates are associated with the gene-specific transcription factor Ascl1, the core factor TBP2, and the polymerase II (Pol-II) ser5 C-terminal domain (CTD) phosphorylated form, while Pol-II ser2 CTD phosphorylation is restricted to the transcriptionally dominant template. We discover that the active and silent DNA forms are physically separated in the oocyte nucleus through partition into liquid-liquid phase-separated condensates. Altogether, our study proposes a mechanism of transcriptional regulation involving a spatial entrapment of general transcription machinery components to stabilize the active form of a gene in a nondividing cell.


Subject(s)
DNA/genetics , Gene Expression Regulation , Oocytes/metabolism , Transcription, Genetic , Animals , Cell Differentiation , DNA/metabolism , Humans , Oocytes/cytology , Phosphorylation , RNA Polymerase II/metabolism , Templates, Genetic , Xenopus
3.
Phys Rev Lett ; 127(19): 198102, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34797132

ABSTRACT

In tissues as diverse as amphibian skin and the human airway, the cilia that propel fluid are grouped in sparsely distributed multiciliated cells (MCCs). We investigate fluid transport in this "mosaic" architecture, with emphasis on the trade-offs that may have been responsible for its evolutionary selection. Live imaging of MCCs in embryos of the frog Xenopus laevis shows that cilia bundles behave as active vortices that produce a flow field accurately represented by a local force applied to the fluid. A coarse-grained model that self-consistently couples bundles to the ambient flow reveals that hydrodynamic interactions between MCCs limit their rate of work so that they best shear the tissue at a finite but low area coverage, a result that mirrors findings for other sparse distributions such as cell receptors and leaf stomata.


Subject(s)
Cilia/physiology , Hydrodynamics , Animals , Humans , Xenopus laevis
4.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34105722

ABSTRACT

Absence of a specialized wound epidermis is hypothesized to block limb regeneration in higher vertebrates. However, the factors preventing its formation in regeneration-incompetent animals are poorly understood. To characterize the endogenous molecular and cellular regulators of specialized wound epidermis formation in Xenopus laevis tadpoles, and the loss of their regeneration competency during development, we used single-cell transcriptomics and ex vivo regenerating limb cultures. Transcriptomic analysis revealed that the specialized wound epidermis is not a novel cell state, but a re-deployment of the apical-ectodermal-ridge (AER) programme underlying limb development. Enrichment of secreted inhibitory factors, including Noggin, a morphogen expressed in developing cartilage/bone progenitor cells, are identified as key inhibitors of AER cell formation in regeneration-incompetent tadpoles. These factors can be overridden by Fgf10, which operates upstream of Noggin and blocks chondrogenesis. These results indicate that manipulation of the extracellular environment and/or chondrogenesis may provide a strategy to restore regeneration potential in higher vertebrates.


Subject(s)
Extremities/growth & development , Regeneration/physiology , Xenopus Proteins/metabolism , Xenopus laevis/physiology , Animals , Carrier Proteins , Cell Cycle , Cell Division , Epidermal Cells , Epidermis , Gene Expression Profiling , Larva , Regeneration/genetics , Transcriptome , Xenopus Proteins/genetics , Xenopus laevis/genetics
5.
Ecol Evol ; 11(9): 3771-3793, 2021 May.
Article in English | MEDLINE | ID: mdl-33976774

ABSTRACT

Biodiversity has undergone a major decline throughout recent decades, particularly in farmland. Agricultural practices are recognized to be an important pressure on farmland biodiversity, and pesticides are suspected to be one of the main causes of this decline in biodiversity. As part of the national plan for reduction of pesticides use (Ecophyto), the French ministry of agriculture launched the 500 ENI (nonintended effects) monitoring program in 2012 in order to assess the unintended effects of agricultural practices, including pesticide use, on biodiversity represented by several taxonomic groups of interest for farmers. This long-term program monitors the biodiversity of nontargeted species (earthworms, plants, coleoptera, and birds), together with a wide range of annual data on agricultural practices (crop rotation, soil tillage, weed control, fertilizers, chemical treatments, etc.). Other parameters (e.g., landscape and climatic characteristics) are also integrated as covariates during the analyses. This monitoring program is expected to improve our understanding of the relative contribution of the different drivers of population and community trends. Here, we present the experience of setting up the 500 ENI network for this ambitious and highly complex monitoring program, as well as the type of data it collects. The issue of data quality control and some first results are discussed. With the aim of being useful to readers who would like to set up similar monitoring schemes, we also address some questions that have arisen following the first five years of the implementation phase of the program.

6.
Nat Commun ; 11(1): 3491, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32661239

ABSTRACT

Sperm contributes genetic and epigenetic information to the embryo to efficiently support development. However, the mechanism underlying such developmental competence remains elusive. Here, we investigated whether all sperm cells have a common epigenetic configuration that primes transcriptional program for embryonic development. Using calibrated ChIP-seq, we show that remodelling of histones during spermiogenesis results in the retention of methylated histone H3 at the same genomic location in most sperm cell. This homogeneously methylated fraction of histone H3 in the sperm genome is maintained during early embryonic replication. Such methylated histone fraction resisting post-fertilisation reprogramming marks developmental genes whose expression is perturbed upon experimental reduction of histone methylation. A similar homogeneously methylated histone H3 fraction is detected in human sperm. Altogether, we uncover a conserved mechanism of paternal epigenetic information transmission to the embryo through the homogeneous retention of methylated histone in a sperm cells population.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Histones/genetics , Histones/metabolism , Male , Spermatogenesis/genetics , Spermatogenesis/physiology , Xenopus
7.
Structure ; 28(6): 674-689.e11, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32375023

ABSTRACT

Centrioles are cylindrical assemblies whose peripheral microtubule array displays a 9-fold rotational symmetry that is established by the scaffolding protein SAS6. Centriole symmetry can be broken by centriole-associated structures, such as the striated fibers in Chlamydomonas that are important for ciliary function. The conserved protein CCDC61/VFL3 is involved in this process, but its exact role is unclear. Here, we show that CCDC61 is a paralog of SAS6. Crystal structures of CCDC61 demonstrate that it contains two homodimerization interfaces that are similar to those found in SAS6, but result in the formation of linear filaments rather than rings. Furthermore, we show that CCDC61 binds microtubules and that residues involved in CCDC61 microtubule binding are important for ciliary function in Chlamydomonas. Together, our findings suggest that CCDC61 and SAS6 functionally diverged from a common ancestor while retaining the ability to scaffold the assembly of basal body-associated structures or centrioles, respectively.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chlamydomonas/physiology , Cilia/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Algal Proteins/chemistry , Algal Proteins/metabolism , Cell Line , Chlamydomonas/classification , Crystallography, X-Ray , HEK293 Cells , Humans , Microtubules/metabolism , Models, Molecular , Phylogeny , Protein Conformation , Protein Domains , Protein Multimerization
8.
Development ; 147(3)2020 02 05.
Article in English | MEDLINE | ID: mdl-31988186

ABSTRACT

Regeneration-competent vertebrates are considered to suppress inflammation faster than non-regenerating ones. Hence, understanding the cellular mechanisms affected by immune cells and inflammation can help develop strategies to promote tissue repair and regeneration. Here, we took advantage of naturally occurring tail regeneration-competent and -incompetent developmental stages of Xenopus tadpoles. We first establish the essential role of the myeloid lineage for tail regeneration in the regeneration-competent tadpoles. We then reveal that upon tail amputation there is a myeloid lineage-dependent change in amputation-induced apoptosis levels, which in turn promotes tissue remodelling, and ultimately leads to the relocalization of the regeneration-organizing cells responsible for progenitor proliferation. These cellular mechanisms failed to be executed in regeneration-incompetent tadpoles. We demonstrate that regeneration incompetency is characterized by inflammatory myeloid cells whereas regeneration competency is associated with reparative myeloid cells. Moreover, treatment of regeneration-incompetent tadpoles with immune-suppressing drugs restores myeloid lineage-controlled cellular mechanisms. Collectively, our work reveals the effects of differential activation of the myeloid lineage on the creation of a regeneration-permissive environment and could be further exploited to devise strategies for regenerative medicine purposes.


Subject(s)
Cell Lineage/physiology , Myeloid Cells/physiology , Regeneration/physiology , Tail/physiology , Xenopus laevis/physiology , Animals , Apoptosis/drug effects , Extracellular Matrix/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Immunosuppressive Agents/pharmacology , Larva/physiology , Regeneration/drug effects , Regenerative Medicine/methods
9.
Cell Rep ; 24(2): 304-311, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29996092

ABSTRACT

Oocytes have a remarkable ability to reactivate silenced genes in somatic cells. However, it is not clear how the chromatin architecture of somatic cells affects this transcriptional reprogramming. Here, we investigated the relationship between the chromatin opening and transcriptional activation. We reveal changes in chromatin accessibility and their relevance to transcriptional reprogramming after transplantation of somatic nuclei into Xenopus oocytes. Genes that are silenced, but have pre-existing open transcription start sites in donor cells, are prone to be activated after nuclear transfer, suggesting that the chromatin signature of somatic nuclei influences transcriptional reprogramming. There are also activated genes associated with new open chromatin sites, and transcription factors in oocytes play an important role in transcriptional reprogramming from such genes. Finally, we show that genes resistant to reprogramming are associated with closed chromatin configurations. We conclude that chromatin accessibility is a central factor for successful transcriptional reprogramming in oocytes.


Subject(s)
Cellular Reprogramming/genetics , Chromatin/metabolism , Oocytes/metabolism , Transcription, Genetic , Animals , Fibroblasts/cytology , Fibroblasts/transplantation , Mice , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , Transcription Factors/metabolism , Transcription Initiation Site , Transcriptional Activation/genetics , Transposases/metabolism , Xenopus laevis/metabolism
10.
Cell Stem Cell ; 21(1): 135-143.e6, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28366589

ABSTRACT

Vertebrate eggs can induce the nuclear reprogramming of somatic cells to enable production of cloned animals. Nuclear reprogramming is relatively inefficient, and the development of the resultant embryos is frequently compromised, in part due to the inappropriate expression of genes previously active in the donor nucleus. Here, we identify H3K4 methylation as a major epigenetic roadblock that limits transcriptional reprogramming and efficient nuclear transfer (NT). Widespread expression of donor-cell-specific genes was observed in inappropriate cell types in NT embryos, limiting their developmental capacity. The expression of these genes in reprogrammed embryos arises from epigenetic memories of a previously active transcriptional state in donor cells that is characterized by high H3K4 methylation. Reducing H3K4 methylation had little effect on gene expression in donor cells, but it substantially improved transcriptional reprogramming and development of NT embryos. These results show that H3K4 methylation imposes a barrier to efficient nuclear reprogramming and suggest approaches for improving reprogramming strategies.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Histones/metabolism , Nuclear Transfer Techniques , Xenopus Proteins/metabolism , Animals , Female , Histones/genetics , Male , Methylation , Mice , Xenopus Proteins/genetics , Xenopus laevis
11.
Biol Open ; 6(4): 415-424, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28412714

ABSTRACT

Animal cloning has been achieved in many species by transplanting differentiated cell nuclei to unfertilized oocytes. However, the low efficiencies of cloning have remained an unresolved issue. Here we find that the combination of two small molecules, trichostatin A (TSA) and vitamin C (VC), under culture condition with bovine serum albumin deionized by ion-exchange resins, dramatically improves the cloning efficiency in mice and 15% of cloned embryos develop to term by means of somatic cell nuclear transfer (SCNT). The improvement was not observed by adding the non-treated, rather than deionized, bovine serum. RNA-seq analyses of SCNT embryos at the two-cell stage revealed that the treatment with TSA and VC resulted in the upregulated expression of previously identified reprogramming-resistant genes. Moreover, the expression of early-embryo-specific retroelements was upregulated by the TSA and VC treatment. The enhanced gene expression was relevant to the VC-mediated reduction of histone H3 lysine 9 methylation in SCNT embryos. Our study thus shows a simply applicable method to greatly improve mouse cloning efficiency, and furthers our understanding of how somatic nuclei acquire totipotency.

12.
Mol Cell ; 65(5): 873-884.e8, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28257702

ABSTRACT

Understanding the mechanism of resistance of genes to reactivation will help improve the success of nuclear reprogramming. Using mouse embryonic fibroblast nuclei with normal or reduced DNA methylation in combination with chromatin modifiers able to erase H3K9me3, H3K27me3, and H2AK119ub1 from transplanted nuclei, we reveal the basis for resistance of genes to transcriptional reprogramming by oocyte factors. A majority of genes is affected by more than one type of treatment, suggesting that resistance can require repression through multiple epigenetic mechanisms. We classify resistant genes according to their sensitivity to 11 chromatin modifier combinations, revealing the existence of synergistic as well as adverse effects of chromatin modifiers on removal of resistance. We further demonstrate that the chromatin modifier USP21 reduces resistance through its H2AK119 deubiquitylation activity. Finally, we provide evidence that H2A ubiquitylation also contributes to resistance to transcriptional reprogramming in mouse nuclear transfer embryos.


Subject(s)
Cell Nucleus/metabolism , Cellular Reprogramming , Chromatin/metabolism , DNA Methylation , Epigenesis, Genetic , Histones/metabolism , Nuclear Transfer Techniques , Transcription, Genetic , Animals , Animals, Genetically Modified , Cell Line , Chromatin/genetics , Chromatin Assembly and Disassembly , Cloning, Molecular , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Oocytes , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Xenopus laevis
13.
Int J Mol Sci ; 18(4)2017 Mar 24.
Article in English | MEDLINE | ID: mdl-28338624

ABSTRACT

Membrane microdomains or "lipid rafts" have emerged as essential functional modules of the cell, critical for the regulation of growth factor receptor-mediated responses. Herein we describe the dichotomy between caveolin-1 and caveolin-2, structural and regulatory components of microdomains, in modulating proliferation and differentiation. Caveolin-2 potentiates while caveolin-1 inhibits nerve growth factor (NGF) signaling and subsequent cell differentiation. Caveolin-2 does not appear to impair NGF receptor trafficking but elicits prolonged and stronger activation of MAPK (mitogen-activated protein kinase), Rsk2 (ribosomal protein S6 kinase 2), and CREB (cAMP response element binding protein). In contrast, caveolin-1 does not alter initiation of the NGF signaling pathway activation; rather, it acts, at least in part, by sequestering the cognate receptors, TrkA and p75NTR, at the plasma membrane, together with the phosphorylated form of the downstream effector Rsk2, which ultimately prevents CREB phosphorylation. The non-phosphorylatable caveolin-1 serine 80 mutant (S80V), no longer inhibits TrkA trafficking or subsequent CREB phosphorylation. MC192, a monoclonal antibody towards p75NTR that does not block NGF binding, prevents exit of both NGF receptors (TrkA and p75NTR) from lipid rafts. The results presented herein underline the role of caveolin and receptor signaling complex interplay in the context of neuronal development and tumorigenesis.


Subject(s)
Caveolin 1/metabolism , Cell Nucleus/metabolism , Membrane Microdomains/metabolism , Nerve Growth Factor/pharmacology , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , CREB-Binding Protein/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 2/antagonists & inhibitors , Caveolin 2/genetics , Caveolin 2/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Mice , Nerve Tissue Proteins , PC12 Cells , Phosphorylation/drug effects , Protein Binding , Protein Transport/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/chemistry , Receptor, trkA/immunology , Receptor, trkA/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/immunology , Receptors, Nerve Growth Factor/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
14.
Genome Res ; 26(8): 1034-46, 2016 08.
Article in English | MEDLINE | ID: mdl-27034506

ABSTRACT

For a long time, it has been assumed that the only role of sperm at fertilization is to introduce the male genome into the egg. Recently, ideas have emerged that the epigenetic state of the sperm nucleus could influence transcription in the embryo. However, conflicting reports have challenged the existence of epigenetic marks on sperm genes, and there are no functional tests supporting the role of sperm epigenetic marking on embryonic gene expression. Here, we show that sperm is epigenetically programmed to regulate embryonic gene expression. By comparing the development of sperm- and spermatid-derived frog embryos, we show that the programming of sperm for successful development relates to its ability to regulate transcription of a set of developmentally important genes. During spermatid maturation into sperm, these genes lose H3K4me2/3 and retain H3K27me3 marks. Experimental removal of these epigenetic marks at fertilization de-regulates gene expression in the resulting embryos in a paternal chromatin-dependent manner. This demonstrates that epigenetic instructions delivered by the sperm at fertilization are required for correct regulation of gene expression in the future embryos. The epigenetic mechanisms of developmental programming revealed here are likely to relate to the mechanisms involved in transgenerational transmission of acquired traits. Understanding how parental experience can influence development of the progeny has broad potential for improving human health.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Spermatozoa/metabolism , Animals , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/biosynthesis , Histones , Humans , Male , Ranidae/genetics , Ranidae/growth & development , Spermatids/growth & development , Spermatids/metabolism , Spermatozoa/growth & development
15.
PLoS One ; 10(11): e0142946, 2015.
Article in English | MEDLINE | ID: mdl-26580070

ABSTRACT

Recent advances in genome editing using programmable nucleases have revolutionized gene targeting in various organisms. Successful gene knock-out has been shown in Xenopus, a widely used model organism, although a system enabling less mosaic knock-out in founder embryos (F0) needs to be explored in order to judge phenotypes in the F0 generation. Here, we injected modified highly active transcription activator-like effector nuclease (TALEN) mRNA to oocytes at the germinal vesicle (GV) stage, followed by in vitro maturation and intracytoplasmic sperm injection, to achieve a full knock-out in F0 embryos. Unlike conventional injection methods to fertilized embryos, the injection of TALEN mRNA into GV oocytes allows expression of nucleases before fertilization, enabling them to work from an earlier stage. Using this procedure, most of developed embryos showed full knock-out phenotypes of the pigmentation gene tyrosinase and/or embryonic lethal gene pax6 in the founder generation. In addition, our method permitted a large 1 kb deletion. Thus, we describe nearly complete gene knock-out phenotypes in Xenopus laevis F0 embryos. The presented method will help to accelerate the production of knock-out frogs since we can bypass an extra generation of about 1 year in Xenopus laevis. Meantime, our method provides a unique opportunity to rapidly test the developmental effects of disrupting those genes that do not permit growth to an adult able to reproduce. In addition, the protocol shown here is considerably less invasive than the previously used host transfer since our protocol does not require surgery. The experimental scheme presented is potentially applicable to other organisms such as mammals and fish to resolve common issues of mosaicism in founders.


Subject(s)
Deoxyribonucleases/genetics , Founder Effect , Gene Knockout Techniques/methods , RNA, Messenger/genetics , Xenopus laevis/genetics , Animals , Base Sequence , Deoxyribonucleases/metabolism , Embryo, Nonmammalian , Eye Proteins/genetics , Female , Fertilization , Gene Expression Regulation, Developmental , Genes, Lethal , Homeodomain Proteins/genetics , Male , Microinjections , Molecular Sequence Data , Monophenol Monooxygenase/deficiency , Monophenol Monooxygenase/genetics , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Phenotype , RNA, Messenger/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , Sequence Alignment , Sperm Injections, Intracytoplasmic , Transcriptional Activation , Xenopus laevis/embryology
16.
Sci Rep ; 5: 14236, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26387861

ABSTRACT

Transposable elements in the genome are generally silenced in differentiated somatic cells. However, increasing evidence indicates that some of them are actively transcribed in early embryos and the proper regulation of retrotransposon expression is essential for normal development. Although their developmentally regulated expression has been shown, the mechanisms controlling retrotransposon expression in early embryos are still not well understood. Here, we observe a dynamic expression pattern of retrotransposons with three out of ten examined retrotransposons (1a11, λ-olt 2-1 and xretpos(L)) being transcribed solely during early embryonic development. We also identified a transcript that contains the long terminal repeat (LTR) of λ-olt 2-1 and shows a similar expression pattern to λ-olt 2-1 in early Xenopus embryos. All three retrotransposons are transcribed by RNA polymerase II. Although their expression levels decline during development, the LTRs are marked by histone H3 lysine 4 trimethylation. Furthermore, retrotransposons, especially λ-olt 2-1, are enriched with histone H3 lysine 9 trimethylation (H3K9me3) when their expression is repressed. Overexpression of lysine-specific demethylase 4d removes H3K9me3 marks from Xenopus embryos and inhibits the repression of λ-olt 2-1 after gastrulation. Thus, our study shows that H3K9me3 is important for silencing the developmentally regulated retrotransposon in Xenopus laevis.


Subject(s)
Gene Silencing , Histones/metabolism , Retroelements/genetics , Xenopus laevis/embryology , Animals , Base Sequence , Cell Differentiation/genetics , Histone Demethylases/metabolism , Methylation , RNA Polymerase II/metabolism , Sequence Analysis, RNA , Sequence Homology, Nucleic Acid , Transcription, Genetic/genetics
17.
Methods Mol Biol ; 1222: 71-82, 2015.
Article in English | MEDLINE | ID: mdl-25287339

ABSTRACT

Germinal vesicle of stage V-VI Xenopus Laevis oocytes (at the prophase I stage of meiosis) can be used to transplant mammalian nuclei. In this type of interspecies nuclear transfer no cell division occurs and no new cell types are generated. However, the transplanted nuclei undergo extensive transcriptional reprogramming. Here, it is first explained how to carry out transplantation of multiple mammalian cell nuclei to Xenopus oocytes. It is then described how to perform RT-qPCR, Western Blot, Chromatin Immunoprecipitation, and live imaging analysis to monitor transcriptional reprogramming of the nuclei transplanted to oocytes.


Subject(s)
Cellular Reprogramming , Image Processing, Computer-Assisted/methods , Nuclear Transfer Techniques , Oocytes/cytology , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Blotting, Western , Cell Nucleus , Chromatin Immunoprecipitation , Female , Image Processing, Computer-Assisted/instrumentation , Microinjections/instrumentation , Microinjections/methods , Nuclear Transfer Techniques/instrumentation , Xenopus laevis
18.
Int J Mol Sci ; 15(9): 16719-40, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25244019

ABSTRACT

Spermatozoa are more efficient at supporting normal embryonic development than spermatids, their immature, immediate precursors. This suggests that the sperm acquires the ability to support embryonic development during spermiogenesis (spermatid to sperm maturation). Here, using Xenopus laevis as a model organism, we performed 2-D Fluorescence Difference Gel Electrophoresis (2D-DIGE) and mass spectrometry analysis of differentially expressed proteins between sperm and spermatids in order to identify factors that could be responsible for the efficiency of the sperm to support embryonic development. Furthermore, benefiting from the availability of egg extracts in Xenopus, we also tested whether the chromatin of sperm could attract different egg factors compared to the chromatin of spermatids. Our analysis identified: (1) several proteins which were present exclusively in sperm; but not in spermatid nuclei and (2) numerous egg proteins binding to the sperm (but not to the spermatid chromatin) after incubation in egg extracts. Amongst these factors we identified many chromatin-associated proteins and transcriptional repressors. Presence of transcriptional repressors binding specifically to sperm chromatin could suggest its preparation for the early embryonic cell cycles, during which no transcription is observed and suggests that sperm chromatin has a unique protein composition, which facilitates the recruitment of egg chromatin remodelling factors. It is therefore likely that the acquisition of these sperm-specific factors during spermiogenesis makes the sperm chromatin suitable to interact with the maternal factors and, as a consequence, to support efficient embryonic development.


Subject(s)
Chromatin/metabolism , Egg Proteins/metabolism , Nuclear Proteins/metabolism , Sperm-Ovum Interactions , Spermatids/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , Xenopus Proteins/metabolism , Animals , Chromatin Assembly and Disassembly , Electrophoresis, Polyacrylamide Gel , Female , Immunoblotting , Male , Mass Spectrometry , Nuclear Proteins/isolation & purification , Protein Binding , Protein Interaction Mapping , Protein Isoforms , Tissue Extracts , Xenopus Proteins/isolation & purification , Xenopus laevis/metabolism
19.
PLoS Biol ; 12(7): e1001914, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25072650

ABSTRACT

Cell differentiation is remarkably stable but can be reversed by somatic cell nuclear transfer, cell fusion, and iPS. Nuclear transfer to amphibian oocytes provides a special opportunity to test transcriptional reprogramming without cell division. We show here that, after nuclear transfer to amphibian oocytes, mitotic chromatin is reprogrammed up to 100 times faster than interphase nuclei. We find that, as cells traverse mitosis, their genes pass through a temporary phase of unusually high responsiveness to oocyte reprogramming factors (mitotic advantage). Mitotic advantage is not explained by nuclear penetration, DNA modifications, histone acetylation, phosphorylation, methylation, nor by salt soluble chromosomal proteins. Our results suggest that histone H2A deubiquitination may account, at least in part, for the acquisition of mitotic advantage. They support the general principle that a temporary access of cytoplasmic factors to genes during mitosis may facilitate somatic cell nuclear reprogramming and the acquisition of new cell fates in normal development.


Subject(s)
Cellular Reprogramming , Chromatin/metabolism , Mitosis/physiology , Transcription, Genetic , Amphibians , Animals , Cell Line , Histones/metabolism , Mice , Nuclear Transfer Techniques , Oocytes/metabolism
20.
Mol Cell ; 55(4): 524-36, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25066233

ABSTRACT

Nuclear transfer to oocytes is an efficient way to transcriptionally reprogram somatic nuclei, but its mechanisms remain unclear. Here, we identify a sequence of molecular events that leads to rapid transcriptional reprogramming of somatic nuclei after transplantation to Xenopus oocytes. RNA-seq analyses reveal that reprogramming by oocytes results in a selective switch in transcription toward an oocyte rather than pluripotent type, without requiring new protein synthesis. Time-course analyses at the single-nucleus level show that transcriptional reprogramming is induced in most transplanted nuclei in a highly hierarchical manner. We demonstrate that an extensive exchange of somatic- for oocyte-specific factors mediates reprogramming and leads to robust oocyte RNA polymerase II binding and phosphorylation on transplanted chromatin. Moreover, genome-wide binding of oocyte-specific linker histone B4 supports its role in transcriptional reprogramming. Thus, our study reveals the rapid, abundant, and stepwise loading of oocyte-specific factors onto somatic chromatin as important determinants for successful reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Chromatin/metabolism , Histones/physiology , Oocytes/metabolism , Xenopus/embryology , Animals , Cells, Cultured , Cellular Reprogramming/physiology , Genome , Mice , Nuclear Transfer Techniques , Organ Specificity , RNA/genetics , Sequence Analysis, RNA , Xenopus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL