64 results on '"Pasque V"'
Search Results
2. Mammalian nuclear transplantation to Germinal Vesicle stage Xenopus oocytes – A method for quantitative transcriptional reprogramming
- Author
-
Halley-Stott, R.P., Pasque, V., Astrand, C., Miyamoto, K., Simeoni, I., Jullien, J., and Gurdon, J.B.
- Published
- 2010
- Full Text
- View/download PDF
3. Hierarchical molecular events driven by oocyte-specific factors lead to rapid and extensive reprogramming
- Author
-
Jullien, J., Miyamoto, K., Pasque, V., Allen, G. E., Bradshaw, C. R., Garrett, N. J., Halley-Stott, R. P., Kimura, Hiroshi, Ohsumi, K., and Gurdon, J. B.
- Subjects
Mice ,Nuclear Transfer Techniques ,Oocytes/*metabolism ,Genome ,Chromatin/*metabolism ,Organ Specificity ,Sequence Analysis, RNA ,Cellular Reprogramming/*genetics/physiology ,Animals ,RNA/genetics ,Xenopus/*embryology/genetics ,Cells, Cultured ,Histones/*physiology - Published
- 2014
4. The function of the Egr1 transcription factor in cartilage formation and adaptation to microgravity in zebrafish,Danio rerio
- Author
-
Muller, M., primary, Dalcq, J., additional, Aceto, J., additional, Larbuisson, A., additional, Pasque, V., additional, Nourizadeh-Lilladadi, R., additional, Alestrom, P., additional, and Martial, J. A., additional
- Published
- 2010
- Full Text
- View/download PDF
5. Efficiencies and Mechanisms of Nuclear Reprogramming
- Author
-
Pasque, V., primary, Miyamoto, K., additional, and Gurdon, J. B., additional
- Published
- 2010
- Full Text
- View/download PDF
6. The function of the Egr1 transcription factor in cartilage formation and adaptation to microgravity in zebrafish, Danio rerio.
- Author
-
Muller, M., Dalcq, J., Aceto, J., Larbuisson, A., Pasque, V., Nourizadeh-Lilladadi, R., Alestrom, P., and Martial, J. A.
- Subjects
TRANSCRIPTION factors ,ZEBRA danio ,CYPRINIDAE ,GENE expression ,FISH genetics - Abstract
Osteoporosis is one of the major concerns for an ageing human population and for passengers on long-term space flights. Teleosts represent a potentially interesting alternative for studying bone physiology. In zebrafish ( Danio rerio), the cartilaginous elements that form the pharyngeal arches derive from cranial neural crest cells, whose proper patterning and morphogenesis require reciprocal interactions with other tissue types such as pharyngeal endoderm, ectoderm and mesoderm. We show how the zebrafish can be used to study the function of signal transduction pathways, such as the Fgf pathway, or that of particular genes, such as the zinc finger transcription factor Egr1, in pharyngeal skeleton formation and maintenance. We investigate the changes caused by microgravity and chemical treatments on zebrafish. We analyze early gene expression modification using whole genome microarray experiments and the long-term consequences by staining bone structures. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
7. Gene Resistance to Transcriptional Reprogramming following Nuclear Transfer Is Directly Mediated by Multiple Chromatin-Repressive Pathways
- Author
-
Jullien, J, Vodnala, M, Pasque, V, Oikawa, M, Miyamoto, K, Allen, G, David, SA, Brochard, V, Wang, S, Bradshaw, C, Koseki, H, Sartorelli, V, Beaujean, N, and Gurdon, J
- Subjects
resistance ,nuclear transfer ,transcriptional reprogramming ,chromatin ,oocyte ,xenopus ,epigenetic ,3. Good health - 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.
8. Histone variant macroH2A1 regulates synchronous firing of replication origins in the inactive X chromosome.
- Author
-
Arroyo M, Casas-Delucchi CS, Pabba MK, Prorok P, Pradhan SK, Rausch C, Lehmkuhl A, Maiser A, Buschbeck M, Pasque V, Bernstein E, Luck K, and Cardoso MC
- Subjects
- Animals, Chromosomes, Human, X genetics, DNA Helicases metabolism, DNA Helicases genetics, Minichromosome Maintenance Complex Component 3 genetics, Minichromosome Maintenance Complex Component 3 metabolism, Protein Isoforms genetics, Protein Isoforms metabolism, X Chromosome Inactivation genetics, Mice, DNA Replication genetics, Histones metabolism, Histones genetics, Nucleosomes metabolism, Nucleosomes genetics, Replication Origin genetics
- Abstract
MacroH2A has been linked to transcriptional silencing, cell identity, and is a hallmark of the inactive X chromosome (Xi). However, it remains unclear whether macroH2A plays a role in DNA replication. Using knockdown/knockout cells for each macroH2A isoform, we show that macroH2A-containing nucleosomes slow down replication progression rate in the Xi reflecting the higher nucleosome stability. Moreover, macroH2A1, but not macroH2A2, regulates the number of nano replication foci in the Xi, and macroH2A1 downregulation increases DNA loop sizes corresponding to replicons. This relates to macroH2A1 regulating replicative helicase loading during G1 by interacting with it. We mapped this interaction to a phenylalanine in macroH2A1 that is not conserved in macroH2A2 and the C-terminus of Mcm3 helicase subunit. We propose that macroH2A1 enhances the licensing of pre-replication complexes via DNA helicase interaction and loading onto the Xi., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
9. Early human development and stem cell-based human embryo models.
- Author
-
Shahbazi MN and Pasque V
- Subjects
- Humans, Models, Biological, Embryo, Mammalian cytology, Cell Lineage, Stem Cells cytology, Animals, Embryonic Development
- Abstract
The use of stem cells to model the early human embryo promises to transform our understanding of developmental biology and human reproduction. In this review, we present our current knowledge of the first 2 weeks of human embryo development. We first focus on the distinct cell lineages of the embryo and the derivation of stem cell lines. We then discuss the intercellular crosstalk that guides early embryo development and how this crosstalk is recapitulated in vitro to generate stem cell-based embryo models. We highlight advances in this fast-developing field, discuss current limitations, and provide a vision for the future., Competing Interests: Declaration of interests The KU Leuven, Belgium, has filed patent application PCT/EP2023/073949 describing the protocols for inducing ExM cells using naive human PSCs. V.P. is one of the inventors of this patent., (Copyright © 2024 MRC Laboratory of Molecular Biology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Unraveling hallmark suitability for staging pre- and post-implantation stem cell models.
- Author
-
Onfray C, Chevolleau S, Moinard E, Girard O, Mahadik K, Allsop R, Georgolopoulos G, Lavigne R, Renoult O, Aksoy I, Lemaitre E, Hulin P, Ouimette JF, Fréour T, Pecqueur C, Pineau C, Pasque V, Rougeulle C, and David L
- Subjects
- Humans, DNA Methylation, Pluripotent Stem Cells metabolism, Pluripotent Stem Cells cytology, Models, Biological, Embryo Implantation, Cell Differentiation, Epigenesis, Genetic, Transcriptome genetics, Proteomics methods, Trophoblasts metabolism, Trophoblasts cytology
- Abstract
The advent of novel 2D and 3D models for human development, including trophoblast stem cells and blastoids, has expanded opportunities for investigating early developmental events, gradually illuminating the enigmatic realm of human development. While these innovations have ushered in new prospects, it has become essential to establish well-defined benchmarks for the cell sources of these models. We aimed to propose a comprehensive characterization of pluripotent and trophoblastic stem cell models by employing a combination of transcriptomic, proteomic, epigenetic, and metabolic approaches. Our findings reveal that extended pluripotent stem cells share many characteristics with primed pluripotent stem cells, with the exception of metabolic activity. Furthermore, our research demonstrates that DNA hypomethylation and high metabolic activity define trophoblast stem cells. These results underscore the necessity of considering multiple hallmarks of pluripotency rather than relying on a single criterion. Multiplying hallmarks alleviate stage-matching bias., Competing Interests: Declaration of interests The authors declare no conflict of interest., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Spatially Self-Organized Three-Dimensional Neural Concentroid as a Novel Reductionist Humanized Model to Study Neurovascular Development.
- Author
-
Chai YC, To SK, Simorgh S, Zaunz S, Zhu Y, Ahuja K, Lemaitre A, Ramezankhani R, van der Veer BK, Wierda K, Verhulst S, van Grunsven LA, Pasque V, and Verfaillie C
- Subjects
- Humans, Neurogenesis physiology, Cell Differentiation physiology, Brain, Endothelial Cells physiology, Pluripotent Stem Cells
- Abstract
Although human pluripotent stem cell (PSC)-derived brain organoids have enabled researchers to gain insight into human brain development and disease, these organoids contain solely ectodermal cells and are not vascularized as occurs during brain development. Here it is created less complex and more homogenous large neural constructs starting from PSC-derived neuroprogenitor cells (NPC), by fusing small NPC spheroids into so-called concentroids. Such concentroids consisted of a pro-angiogenic core, containing neuronal and outer radial glia cells, surrounded by an astroglia-dense outer layer. Incorporating PSC-derived endothelial cells (EC) around and/or in the concentroids promoted vascularization, accompanied by differential outgrowth and differentiation of neuronal and astroglia cells, as well as the development of ectodermal-derived pericyte-like mural cells co-localizing with EC networks. Single nucleus transcriptomic analysis revealed an enhanced neural cell subtype maturation and diversity in EC-containing concentroids, which better resemble the fetal human brain compared to classical organoids or NPC-only concentroids. This PSC-derived "vascularized" concentroid brain model will facilitate the study of neurovascular/blood-brain barrier development, neural cell migration, and the development of effective in vitro vascularization strategies of brain mimics., (© 2023 The Authors. Advanced Science published by Wiley-VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
12. Induction of Human Extraembryonic Mesoderm Cells from Naive Pluripotent Stem Cells.
- Author
-
Panda A, Pham TXA, Khodeer S, and Pasque V
- Subjects
- Animals, Humans, Embryo, Mammalian, Embryonic Development, Primates, Cell Differentiation, Mesoderm, Pluripotent Stem Cells
- Abstract
The human extraembryonic mesoderm (EXM) is an important tissue in the postimplantation embryo which is specified before gastrulation in primates but not in rodents. EXM is mesenchymal and plays an important role in embryogenesis, including early erythropoiesis, and provides mechanical support to the developing embryo. Recently, it has been shown that self-renewing extraembryonic mesoderm cells (EXMCs) can be modeled in vitro by using human naive pluripotent stem cells. Here, we present a detailed step-by-step protocol to induce EXMCs from naive pluripotent stem cells in vitro., (© 2023. Springer Science+Business Media, LLC.)
- Published
- 2024
- Full Text
- View/download PDF
13. Evaluation of Stem-Cell Embryo Models by Integration with a Human Embryo Single-Cell Transcriptome Atlas.
- Author
-
To SK, Balaton B, and Pasque V
- Subjects
- Humans, Blastocyst, Trophoblasts, Stem Cells, Single-Cell Analysis, Transcriptome, Embryo, Mammalian
- Abstract
Single-cell RNA sequencing (scRNA-seq) revolutionized our understanding of the molecular processes of early development and provided us with the means to capture biological heterogeneity and assess the cellular composition in early embryos. Comparative analysis of the transcriptional landscapes of embryos with single-cell resolution allows us to better understand and improve stem-cell-based embryo models. However, proper comparison between different single-cell datasets acquired by different laboratories and through different technologies is imperative for adequate analysis and findings. In this chapter, we focus on the analysis of human blastoids, which model the blastocyst, and their integrative analysis with human embryo datasets and a 2D in vitro early development model system dataset, which models epiblast, extraembryonic mesoderm, and trophoblast cells., (© 2023. Springer Science+Business Media, LLC.)
- Published
- 2024
- Full Text
- View/download PDF
14. A facile method to generate cerebral organoids from human pluripotent stem cells.
- Author
-
Simorgh S, Mousavi SA, To SK, Pasque V, Wierda K, Vervliet T, Yeganeh M, Pooyan P, Chai YC, Verfaillie C, and Baharvand H
- Abstract
Human cerebral organoids (COs) are self-organizing three-dimensional (3D) neural structures that provide a human-specific platform to study the cellular and molecular processes that underlie different neurological events. The first step of CO generation from human pluripotent stem cells (hPSCs) is neural induction, which is an in vitro simulation of neural ectoderm development. Several signaling pathways cooperate during neural ectoderm development and in vitro differentiation of hPSCs toward neural cell lineages is also affected by them. In this study, we considered some of the known sources of these variable signaling cues arising from cell culture media components and sought to modulate their effects by applying a comprehensive combination of small molecules and growth factors for CO generation. Histological analysis demonstrated that these COs recapitulate the neural progenitor zone and early cortical layer organization, containing different types of neuronal and glial cells which was in accordance with single-nucleus transcriptome profiling results. Moreover, patch clamp and intracellular Ca
2+ dynamic studies demonstrated that the COs behave as a functional neural network. Thus, this method serves as a facile protocol for generating hPSC-derived COs that faithfully mimic the features of their in vivo counterparts in the developing human brain. See also Figure 1(Fig. 1)., Competing Interests: The authors declare that they have no conflict of interest., (Copyright © 2023 Simorgh et al.)- Published
- 2023
- Full Text
- View/download PDF
15. Complete human day 14 post-implantation embryo models from naive ES cells.
- Author
-
Oldak B, Wildschutz E, Bondarenko V, Comar MY, Zhao C, Aguilera-Castrejon A, Tarazi S, Viukov S, Pham TXA, Ashouokhi S, Lokshtanov D, Roncato F, Ariel E, Rose M, Livnat N, Shani T, Joubran C, Cohen R, Addadi Y, Chemla M, Kedmi M, Keren-Shaul H, Pasque V, Petropoulos S, Lanner F, Novershtern N, and Hanna JH
- Subjects
- Humans, Fertilization, Gastrulation, Germ Layers cytology, Germ Layers embryology, Trophoblasts cytology, Yolk Sac cytology, Yolk Sac embryology, Giant Cells cytology, Embryo Implantation, Embryo, Mammalian cytology, Embryo, Mammalian embryology, Embryonic Development, Human Embryonic Stem Cells cytology
- Abstract
The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation
1 . Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2 . Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3 . Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4 . Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
16. Having a blast(oid): Modeling human embryo peri-implantation development with blastoids.
- Author
-
Moinard E, Pasque V, and David L
- Subjects
- Humans, Embryo, Mammalian, Stem Cells, Embryo Implantation, Embryonic Development
- Abstract
Studying human embryo development, most particularly around the time of implantation, is a challenge, yet it is necessary to improve assisted reproduction techniques. In this issue, Yu et al.
1 and Karvas et al.2 improve integrated stem cell models, called blastoids, to model the peri-implantation human embryo., Competing Interests: Declaration of interests V.P. submitted (through KU Leuven) a patent application relevant to the generation of extraembryonic mesoderm from human naive pluripotent stem cells, filed on August 31, 2022 (EP22193280.9)., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
17. Inferring regulators of cell identity in the human adult pancreas.
- Author
-
Vanheer L, Fantuzzi F, To SK, Schiavo A, Van Haele M, Ostyn T, Haesen T, Yi X, Janiszewski A, Chappell J, Rihoux A, Sawatani T, Roskams T, Pattou F, Kerr-Conte J, Cnop M, and Pasque V
- Abstract
Cellular identity during development is under the control of transcription factors that form gene regulatory networks. However, the transcription factors and gene regulatory networks underlying cellular identity in the human adult pancreas remain largely unexplored. Here, we integrate multiple single-cell RNA-sequencing datasets of the human adult pancreas, totaling 7393 cells, and comprehensively reconstruct gene regulatory networks. We show that a network of 142 transcription factors forms distinct regulatory modules that characterize pancreatic cell types. We present evidence that our approach identifies regulators of cell identity and cell states in the human adult pancreas. We predict that HEYL, BHLHE41 and JUND are active in acinar, beta and alpha cells, respectively, and show that these proteins are present in the human adult pancreas as well as in human induced pluripotent stem cell (hiPSC)-derived islet cells. Using single-cell transcriptomics, we found that JUND represses beta cell genes in hiPSC-alpha cells. BHLHE41 depletion induced apoptosis in primary pancreatic islets. The comprehensive gene regulatory network atlas can be explored interactively online. We anticipate our analysis to be the starting point for a more sophisticated dissection of how transcription factors regulate cell identity and cell states in the human adult pancreas., (© The Author(s) 2023. Published by Oxford University Press on behalf of NAR Genomics and Bioinformatics.)
- Published
- 2023
- Full Text
- View/download PDF
18. The human embryo selection arena is associated with transposable element activity.
- Author
-
Osnato A, Pasque V, and David L
- Subjects
- Humans, Embryo, Mammalian, DNA Transposable Elements genetics, Blastocyst
- Abstract
Our current understanding of early human development is limited. A study in PLOS Biology found a previously undefined group of cells that diverges from the main lineages and undergo apoptosis through the activity of young transposable elements., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Osnato et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
19. The Wnt/TCF7L1 transcriptional repressor axis drives primitive endoderm formation by antagonizing naive and formative pluripotency.
- Author
-
Athanasouli P, Balli M, De Jaime-Soguero A, Boel A, Papanikolaou S, van der Veer BK, Janiszewski A, Vanhessche T, Francis A, El Laithy Y, Nigro AL, Aulicino F, Koh KP, Pasque V, Cosma MP, Verfaillie C, Zwijsen A, Heindryckx B, Nikolaou C, and Lluis F
- Subjects
- Animals, Female, Mice, Pregnancy, Blastocyst, Cell Differentiation, Germ Layers, Automobile Driving, Endoderm, Transcription Factor 7-Like 1 Protein
- Abstract
Early during preimplantation development and in heterogeneous mouse embryonic stem cells (mESC) culture, pluripotent cells are specified towards either the primed epiblast or the primitive endoderm (PE) lineage. Canonical Wnt signaling is crucial for safeguarding naive pluripotency and embryo implantation, yet the role and relevance of canonical Wnt inhibition during early mammalian development remains unknown. Here, we demonstrate that transcriptional repression exerted by Wnt/TCF7L1 promotes PE differentiation of mESCs and in preimplantation inner cell mass. Time-series RNA sequencing and promoter occupancy data reveal that TCF7L1 binds and represses genes encoding essential naive pluripotency factors and indispensable regulators of the formative pluripotency program, including Otx2 and Lef1. Consequently, TCF7L1 promotes pluripotency exit and suppresses epiblast lineage formation, thereby driving cells into PE specification. Conversely, TCF7L1 is required for PE specification as deletion of Tcf7l1 abrogates PE differentiation without restraining epiblast priming. Taken together, our study underscores the importance of transcriptional Wnt inhibition in regulating lineage specification in ESCs and preimplantation embryo development as well as identifies TCF7L1 as key regulator of this process., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. Modeling human extraembryonic mesoderm cells using naive pluripotent stem cells.
- Author
-
Pham TXA, Panda A, Kagawa H, To SK, Ertekin C, Georgolopoulos G, van Knippenberg SSFA, Allsop RN, Bruneau A, Chui JS, Vanheer L, Janiszewski A, Chappell J, Oberhuemer M, Tchinda RS, Talon I, Khodeer S, Rossant J, Lluis F, David L, Rivron N, Balaton BP, and Pasque V
- Subjects
- Animals, Cell Differentiation, Embryo, Mammalian, Germ Layers, Humans, Mesoderm, Primates, Pluripotent Stem Cells
- Abstract
A hallmark of primate postimplantation embryogenesis is the specification of extraembryonic mesoderm (EXM) before gastrulation, in contrast to rodents where this tissue is formed only after gastrulation. Here, we discover that naive human pluripotent stem cells (hPSCs) are competent to differentiate into EXM cells (EXMCs). EXMCs are specified by inhibition of Nodal signaling and GSK3B, are maintained by mTOR and BMP4 signaling activity, and their transcriptome and epigenome closely resemble that of human and monkey embryo EXM. EXMCs are mesenchymal, can arise from an epiblast intermediate, and are capable of self-renewal. Thus, EXMCs arising via primate-specific specification between implantation and gastrulation can be modeled in vitro. We also find that most of the rare off-target cells within human blastoids formed by triple inhibition (Kagawa et al., 2021) correspond to EXMCs. Our study impacts our ability to model and study the molecular mechanisms of early human embryogenesis and related defects., Competing Interests: Declaration of interests The Institute for Molecular Biotechnology, Austrian Academy of Sciences has filed patent application EP21151455.9 describing the protocols for human blastoid formation. H.K. and N.R. are the inventors of this patent. All other authors declare no competing interests. J.R. is a member of the Cell Stem Cell advisory board., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. Integrated multi-omics reveal polycomb repressive complex 2 restricts human trophoblast induction.
- Author
-
Zijlmans DW, Talon I, Verhelst S, Bendall A, Van Nerum K, Javali A, Malcolm AA, van Knippenberg SSFA, Biggins L, To SK, Janiszewski A, Admiraal D, Knops R, Corthout N, Balaton BP, Georgolopoulos G, Panda A, Bhanu NV, Collier AJ, Fabian C, Allsop RN, Chappell J, Pham TXA, Oberhuemer M, Ertekin C, Vanheer L, Athanasouli P, Lluis F, Deforce D, Jansen JH, Garcia BA, Vermeulen M, Rivron N, Dhaenens M, Marks H, Rugg-Gunn PJ, and Pasque V
- Subjects
- Cell Differentiation genetics, Chromatin genetics, Histones genetics, Humans, Trophoblasts metabolism, Pluripotent Stem Cells, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism
- Abstract
Human naive pluripotent stem cells have unrestricted lineage potential. Underpinning this property, naive cells are thought to lack chromatin-based lineage barriers. However, this assumption has not been tested. Here we define the chromatin-associated proteome, histone post-translational modifications and transcriptome of human naive and primed pluripotent stem cells. Our integrated analysis reveals differences in the relative abundance and activities of distinct chromatin modules. We identify a strong enrichment of polycomb repressive complex 2 (PRC2)-associated H3K27me3 in the chromatin of naive pluripotent stem cells and H3K27me3 enrichment at promoters of lineage-determining genes, including trophoblast regulators. PRC2 activity acts as a chromatin barrier restricting the differentiation of naive cells towards the trophoblast lineage, whereas inhibition of PRC2 promotes trophoblast-fate induction and cavity formation in human blastoids. Together, our results establish that human naive pluripotent stem cells are not epigenetically unrestricted, but instead possess chromatin mechanisms that oppose the induction of alternative cell fates., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
22. Human 8-cell-like cells discovered.
- Author
-
Balaton BP and Pasque V
- Subjects
- Embryo, Mammalian cytology, Humans, Germ Layers cytology, Pluripotent Stem Cells cytology
- Abstract
Human naive pluripotent stem cells have the remarkable ability to generate blastoids comprising trophectoderm, epiblast, and hypoblast-like cells. In this issue, Taubenschmid-Stowers et al. (2022) show that human naive pluripotent stem cell cultures contain cells that resemble the 8-cell human embryo, providing a model to study zygotic genome activation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
23. Enhanced chromatin accessibility contributes to X chromosome dosage compensation in mammals.
- Author
-
Talon I, Janiszewski A, Theeuwes B, Lefevre T, Song J, Bervoets G, Vanheer L, De Geest N, Poovathingal S, Allsop R, Marine JC, Rambow F, Voet T, and Pasque V
- Subjects
- Alleles, Aneuploidy, Animals, Cellular Reprogramming genetics, Gene Regulatory Networks, Induced Pluripotent Stem Cells metabolism, Mice, RNA-Seq, Single-Cell Analysis, Transcription Factors metabolism, Transcription, Genetic, X Chromosome, Chromatin metabolism, X Chromosome Inactivation
- Abstract
Background: Precise gene dosage of the X chromosomes is critical for normal development and cellular function. In mice, XX female somatic cells show transcriptional X chromosome upregulation of their single active X chromosome, while the other X chromosome is inactive. Moreover, the inactive X chromosome is reactivated during development in the inner cell mass and in germ cells through X chromosome reactivation, which can be studied in vitro by reprogramming of somatic cells to pluripotency. How chromatin processes and gene regulatory networks evolved to regulate X chromosome dosage in the somatic state and during X chromosome reactivation remains unclear., Results: Using genome-wide approaches, allele-specific ATAC-seq and single-cell RNA-seq, in female embryonic fibroblasts and during reprogramming to pluripotency, we show that chromatin accessibility on the upregulated mammalian active X chromosome is increased compared to autosomes. We further show that increased accessibility on the active X chromosome is erased by reprogramming, accompanied by erasure of transcriptional X chromosome upregulation and the loss of increased transcriptional burst frequency. In addition, we characterize gene regulatory networks during reprogramming and X chromosome reactivation, revealing changes in regulatory states. Our data show that ZFP42/REX1, a pluripotency-associated gene that evolved specifically in placental mammals, targets multiple X-linked genes, suggesting an evolutionary link between ZFP42/REX1, X chromosome reactivation, and pluripotency., Conclusions: Our data reveal the existence of intrinsic compensatory mechanisms that involve modulation of chromatin accessibility to counteract X-to-Autosome gene dosage imbalances caused by evolutionary or in vitro X chromosome loss and X chromosome inactivation in mammalian cells., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
24. Integrated pseudotime analysis of human pre-implantation embryo single-cell transcriptomes reveals the dynamics of lineage specification.
- Author
-
Meistermann D, Bruneau A, Loubersac S, Reignier A, Firmin J, François-Campion V, Kilens S, Lelièvre Y, Lammers J, Feyeux M, Hulin P, Nedellec S, Bretin B, Castel G, Allègre N, Covin S, Bihouée A, Soumillon M, Mikkelsen T, Barrière P, Chazaud C, Chappell J, Pasque V, Bourdon J, Fréour T, and David L
- Subjects
- Animals, Blastocyst, Cell Lineage genetics, Germ Layers, Humans, Mice, Embryonic Development genetics, Transcriptome genetics
- Abstract
Understanding lineage specification during human pre-implantation development is a gateway to improving assisted reproductive technologies and stem cell research. Here we employ pseudotime analysis of single-cell RNA sequencing (scRNA-seq) data to reconstruct early mouse and human embryo development. Using time-lapse imaging of annotated embryos, we provide an integrated, ordered, and continuous analysis of transcriptomics changes throughout human development. We reveal that human trophectoderm/inner cell mass transcriptomes diverge at the transition from the B2 to the B3 blastocyst stage, just before blastocyst expansion. We explore the dynamics of the fate markers IFI16 and GATA4 and show that they gradually become mutually exclusive upon establishment of epiblast and primitive endoderm fates, respectively. We also provide evidence that NR2F2 marks trophectoderm maturation, initiating from the polar side, and subsequently spreads to all cells after implantation. Our study pinpoints the precise timing of lineage specification events in the human embryo and identifies transcriptomics hallmarks and cell fate markers., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
25. Interleukin-6 is an activator of pituitary stem cells upon local damage, a competence quenched in the aging gland.
- Author
-
Vennekens A, Laporte E, Hermans F, Cox B, Modave E, Janiszewski A, Nys C, Kobayashi H, Malengier-Devlies B, Chappell J, Matthys P, Garcia MI, Pasque V, Lambrechts D, and Vankelecom H
- Subjects
- Animals, Cell Proliferation, Inflammation pathology, Mice, Organoids pathology, Phenotype, Single-Cell Analysis, Transcriptome genetics, Up-Regulation genetics, Aging pathology, Interleukin-6 metabolism, Pituitary Gland pathology, Stem Cells pathology
- Abstract
Stem cells in the adult pituitary are quiescent yet show acute activation upon tissue injury. The molecular mechanisms underlying this reaction are completely unknown. We applied single-cell transcriptomics to start unraveling the acute pituitary stem cell activation process as occurring upon targeted endocrine cell-ablation damage. This stem cell reaction was contrasted with the aging (middle-aged) pituitary, known to have lost damage-repair capacity. Stem cells in the aging pituitary show regressed proliferative activation upon injury and diminished in vitro organoid formation. Single-cell RNA sequencing uncovered interleukin-6 (IL-6) as being up-regulated upon damage, however only in young but not aging pituitary. Administering IL-6 to young mice promptly triggered pituitary stem cell proliferation, while blocking IL-6 or associated signaling pathways inhibited such reaction to damage. By contrast, IL-6 did not generate a pituitary stem cell activation response in aging mice, coinciding with elevated basal IL-6 levels and raised inflammatory state in the aging gland (inflammaging). Intriguingly, in vitro stem cell activation by IL-6 was discerned in organoid culture not only from young but also from aging pituitary, indicating that the aging gland's stem cells retain intrinsic activatability in vivo, likely impeded by the prevailing inflammatory tissue milieu. Importantly, IL-6 supplementation strongly enhanced the growth capability of pituitary stem cell organoids, thereby expanding their potential as an experimental model. Our study identifies IL-6 as a pituitary stem cell activator upon local damage, a competence quenched at aging, concomitant with raised IL-6/inflammatory levels in the older gland. These insights may open the way to interfering with pituitary aging., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
26. Keep the fate: how chromatin regulators safeguard embryonic stem cell identity.
- Author
-
Janiszewski A, Georgolopoulos G, Balli M, Athanasouli P, Lluis F, and Pasque V
- Subjects
- Animals, Cell Differentiation, Cell Lineage genetics, Embryo, Mammalian, Embryonic Stem Cells, Endoderm, Mice, Blastocyst, Chromatin genetics
- Abstract
Segregation of cells that form the embryo from those that produce the surrounding extra-embryonic tissues is critical for early mammalian development, but the regulatory layers governing these first cell fate decisions remain poorly understood. Recent work in The EMBO Journal identifies two chromatin regulators, Hdac3 and Dax1, that synergistically restrict the developmental potential of mouse embryonic stem cells and act as a lineage barrier to primitive endoderm formation., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
27. Evaluating totipotency using criteria of increasing stringency.
- Author
-
Posfai E, Schell JP, Janiszewski A, Rovic I, Murray A, Bradshaw B, Yamakawa T, Pardon T, El Bakkali M, Talon I, De Geest N, Kumar P, To SK, Petropoulos S, Jurisicova A, Pasque V, Lanner F, and Rossant J
- Subjects
- Animals, Blastomeres metabolism, Embryo, Mammalian metabolism, Embryonic Stem Cells metabolism, Female, Gene Expression Profiling, Gene Regulatory Networks, Male, Mice, Pluripotent Stem Cells metabolism, Single-Cell Analysis, Totipotent Stem Cells metabolism, Blastomeres cytology, Cell Differentiation, Cell Lineage genetics, Embryo, Mammalian cytology, Embryonic Stem Cells cytology, Pluripotent Stem Cells cytology, Totipotent Stem Cells cytology
- Abstract
Totipotency is the ability of a single cell to give rise to all of the differentiated cell types that build the conceptus, yet how to capture this property in vitro remains incompletely understood. Defining totipotency relies on a variety of assays of variable stringency. Here, we describe criteria to define totipotency. We explain how distinct criteria of increasing stringency can be used to judge totipotency by evaluating candidate totipotent cell types in mice, including early blastomeres and expanded or extended pluripotent stem cells. Our data challenge the notion that expanded or extended pluripotent states harbour increased totipotent potential relative to conventional embryonic stem cells under in vitro and in vivo conditions.
- Published
- 2021
- Full Text
- View/download PDF
28. New Insights into X-Chromosome Reactivation during Reprogramming to Pluripotency.
- Author
-
Panda A, Zylicz JJ, and Pasque V
- Subjects
- Animals, Cell Differentiation, Chromatin metabolism, Embryonic Development genetics, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, RNA Interference, RNA, Long Noncoding antagonists & inhibitors, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Cellular Reprogramming, X Chromosome physiology
- Abstract
Dosage compensation between the sexes results in one X chromosome being inactivated during female mammalian development. Chromosome-wide transcriptional silencing from the inactive X chromosome (Xi) in mammalian cells is erased in a process termed X-chromosome reactivation (XCR), which has emerged as a paradigm for studying the reversal of chromatin silencing. XCR is linked with germline development and induction of naive pluripotency in the epiblast, and also takes place upon reprogramming somatic cells to induced pluripotency. XCR depends on silencing of the long non-coding RNA (lncRNA) X inactive specific transcript ( Xist) and is linked with the erasure of chromatin silencing. Over the past years, the advent of transcriptomics and epigenomics has provided new insights into the transcriptional and chromatin dynamics with which XCR takes place. However, multiple questions remain unanswered about how chromatin and transcription related processes enable XCR. Here, we review recent work on establishing the transcriptional and chromatin kinetics of XCR, as well as discuss a model by which transcription factors mediate XCR not only via Xist repression, but also by direct targeting of X-linked genes.
- Published
- 2020
- Full Text
- View/download PDF
29. Regulatory Dynamics of Tet1 and Oct4 Resolve Stages of Global DNA Demethylation and Transcriptomic Changes in Reprogramming.
- Author
-
Bartoccetti M, van der Veer BK, Luo X, Khoueiry R, She P, Bajaj M, Xu J, Janiszewski A, Thienpont B, Pasque V, and Koh KP
- Published
- 2020
- Full Text
- View/download PDF
30. Murine iPSC-derived microglia and macrophage cell culture models recapitulate distinct phenotypical and functional properties of classical and alternative neuro-immune polarisation.
- Author
-
Quarta A, Le Blon D, D'aes T, Pieters Z, Hamzei Taj S, Miró-Mur F, Luyckx E, Van Breedam E, Daans J, Goossens H, Dewilde S, Hens N, Pasque V, Planas AM, Hoehn M, Berneman Z, and Ponsaerts P
- Subjects
- Animals, CX3C Chemokine Receptor 1 metabolism, Cell Differentiation physiology, Disease Models, Animal, Female, Induced Pluripotent Stem Cells physiology, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microglia metabolism, Monocytes metabolism, Neuroimmunomodulation immunology, Phenotype, Receptors, CCR2 metabolism, Cell Culture Techniques methods, Induced Pluripotent Stem Cells metabolism, Neuroimmunomodulation physiology
- Abstract
The establishment and validation of reliable induced pluripotent stem cell (iPSC)-derived in vitro models to study microglia and monocyte/macrophage immune function holds great potential for fundamental and translational neuro-immunology research. In this study, we first demonstrate that ramified CX
3 CR1+ iPSC-microglia (cultured within a neural environment) and round-shaped CX3 CR1- iPSC-macrophages can easily be differentiated from newly established murine CX3 CR1eGFP/+ CCR2RFP/+ iPSC lines. Furthermore, we show that obtained murine iPSC-microglia and iPSC-macrophages are distinct cell populations, even though iPSC-macrophages may upregulate CX3 CR1 expression when cultured within a neural environment. Next, we characterized the phenotypical and functional properties of murine iPSC-microglia and iPSC-macrophages following classical and alternative immune polarisation. While iPSC-macrophages could easily be triggered to adopt a classically-activated or alternatively-activated phenotype following, respectively, lipopolysaccharide + interferon γ or interleukin 13 (IL13) stimulation, iPSC-microglia and iPSC-macrophages cultured within a neural environment displayed a more moderate activation profile as characterised by the absence of MHCII expression upon classical immune polarisation and the absence of Ym1 expression upon alternative immune polarisation. Finally, extending our preceding in vivo studies, this striking phenotypical divergence was also observed for resident microglia and infiltrating monocytes within highly inflammatory cortical lesions in CX3 CR1eGFP/+ CCR2RFP/+ mice subjected to middle cerebral arterial occlusion (MCAO) stroke and following IL13-mediated therapeutic intervention thereon. In conclusion, our study demonstrates that the applied murine iPSC-microglia and iPSC-macrophage culture models are able to recapitulate in vivo microglia and monocyte/macrophage ontogeny and corresponding phenotypical/functional properties upon classical and alternative immune polarisation, and therefore represent a valuable in vitro platform to further study and modulate microglia and (infiltrating) monocyte immune responses under neuro-inflammatory conditions within a neural environment., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
31. Tox4 modulates cell fate reprogramming.
- Author
-
Vanheer L, Song J, De Geest N, Janiszewski A, Talon I, Provenzano C, Oh T, Chappell J, and Pasque V
- Subjects
- Animals, Cell Line, Fibroblasts cytology, High Mobility Group Proteins genetics, Induced Pluripotent Stem Cells cytology, Mice, Neural Stem Cells cytology, Cellular Reprogramming, Fibroblasts metabolism, High Mobility Group Proteins metabolism, Induced Pluripotent Stem Cells metabolism, Neural Stem Cells metabolism
- Abstract
Reprogramming to induced pluripotency induces the switch of somatic cell identity to induced pluripotent stem cells (iPSCs). However, the mediators and mechanisms of reprogramming remain largely unclear. To elucidate the mediators and mechanisms of reprogramming, we used a siRNA-mediated knockdown approach for selected candidate genes during the conversion of somatic cells into iPSCs. We identified Tox4 as a novel factor that modulates cell fate through an assay that determined the efficiency of iPSC reprogramming. We found that Tox4 is needed early in reprogramming to efficiently generate early reprogramming intermediates, irrespective of the reprogramming conditions used. Tox4 enables proper exogenous reprogramming factor expression, and the closing and opening of putative somatic and pluripotency enhancers early during reprogramming, respectively. We show that the TOX4 protein assembles into a high molecular form. Moreover, Tox4 is also required for the efficient conversion of fibroblasts towards the neuronal fate, suggesting a broader role of Tox4 in modulating cell fate. Our study reveals Tox4 as a novel transcriptional modulator of cell fate that mediates reprogramming from the somatic state to the pluripotent and neuronal fate.This article has an associated First Person interview with the first author of the paper., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2019. Published by The Company of Biologists Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
32. Dynamic reversal of random X-Chromosome inactivation during iPSC reprogramming.
- Author
-
Janiszewski A, Talon I, Chappell J, Collombet S, Song J, De Geest N, To SK, Bervoets G, Marin-Bejar O, Provenzano C, Vanheer L, Marine JC, Rambow F, and Pasque V
- Subjects
- Animals, Chromatin genetics, Female, Gene Silencing, Genes, X-Linked genetics, Histone Deacetylases genetics, Mice, Transcriptional Activation genetics, X Chromosome genetics, Cellular Reprogramming genetics, Induced Pluripotent Stem Cells cytology, RNA, Long Noncoding genetics, X Chromosome Inactivation genetics
- Abstract
Induction and reversal of chromatin silencing is critical for successful development, tissue homeostasis, and the derivation of induced pluripotent stem cells (iPSCs). X-Chromosome inactivation (XCI) and reactivation (XCR) in female cells represent chromosome-wide transitions between active and inactive chromatin states. Although XCI has long been studied, providing important insights into gene regulation, the dynamics and mechanisms underlying the reversal of stable chromatin silencing of X-linked genes are much less understood. Here, we use allele-specific transcriptomics to study XCR during mouse iPSC reprogramming in order to elucidate the timing and mechanisms of chromosome-wide reversal of gene silencing. We show that XCR is hierarchical, with subsets of genes reactivating early, late, and very late during reprogramming. Early genes are activated before the onset of late pluripotency genes activation. Early genes are located genomically closer to genes that escape XCI, unlike genes reactivating late. Early genes also show increased pluripotency transcription factor (TF) binding. We also reveal that histone deacetylases (HDACs) restrict XCR in reprogramming intermediates and that the severe hypoacetylation state of the inactive X Chromosome (Xi) persists until late reprogramming stages. Altogether, these results reveal the timing of transcriptional activation of monoallelically repressed genes during iPSC reprogramming, and suggest that allelic activation involves the combined action of chromatin topology, pluripotency TFs, and chromatin regulators. These findings are important for our understanding of gene silencing, maintenance of cell identity, reprogramming, and disease., (© 2019 Janiszewski et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
33. Recent Advances in Understanding the Reversal of Gene Silencing During X Chromosome Reactivation.
- Author
-
Talon I, Janiszewski A, Chappell J, Vanheer L, and Pasque V
- Abstract
Dosage compensation between XX female and XY male cells is achieved by a process known as X chromosome inactivation (XCI) in mammals. XCI is initiated early during development in female cells and is subsequently stably maintained in most somatic cells. Despite its stability, the robust transcriptional silencing of XCI is reversible, in the embryo and also in a number of reprogramming settings. Although XCI has been intensively studied, the dynamics, factors, and mechanisms of X chromosome reactivation (XCR) remain largely unknown. In this review, we discuss how new sequencing technologies and reprogramming approaches have enabled recent advances that revealed the timing of transcriptional activation during XCR. We also discuss the factors and chromatin features that might be important to understand the dynamics and mechanisms of the erasure of transcriptional gene silencing on the inactive X chromosome (Xi).
- Published
- 2019
- Full Text
- View/download PDF
34. X-Chromosome Dosage Modulates Multiple Molecular and Cellular Properties of Mouse Pluripotent Stem Cells Independently of Global DNA Methylation Levels.
- Author
-
Song J, Janiszewski A, De Geest N, Vanheer L, Talon I, El Bakkali M, Oh T, and Pasque V
- Subjects
- Animals, Cell Line, Cellular Reprogramming genetics, Chromatin genetics, Dual-Specificity Phosphatases genetics, Epigenesis, Genetic genetics, Epigenomics methods, Female, Induced Pluripotent Stem Cells physiology, Male, Mice, Mitogen-Activated Protein Kinase Phosphatases genetics, Transcriptome genetics, DNA Methylation genetics, Gene Dosage genetics, Pluripotent Stem Cells physiology, X Chromosome genetics
- Abstract
Reprogramming female mouse somatic cells into induced pluripotent stem cells (iPSCs) leads to X-chromosome reactivation. The extent to which increased X-chromosome dosage (X-dosage) in female iPSCs compared with male iPSCs leads to differences in the properties of iPSCs is still unclear. We show that chromatin accessibility in mouse iPSCs is modulated by X-dosage. Specific sets of transcriptional regulator motifs are enriched in chromatin with increased accessibility in XX or XY iPSCs. The transcriptome, growth and pluripotency exit are also modulated by X-dosage in iPSCs. To understand how increased X-dosage modulates the properties of mouse pluripotent stem cells, we used heterozygous deletions of the X-linked gene Dusp9. We show that X-dosage regulates the transcriptome, open chromatin landscape, growth, and pluripotency exit largely independently of global DNA methylation. Our results provide insights into how gene dosage modulates the epigenetic and genetic mechanisms that regulate cell identity., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
35. Dynamics of DNA Methylation Reprogramming Influenced by X Chromosome Dosage in Induced Pluripotent Stem Cells.
- Author
-
Janiszewski A, Song J, Vanheer L, De Geest N, and Pasque V
- Abstract
How the epigenome of one cell type is remodeled during reprogramming into another unrelated type of cell remains unclear. Overexpression of transcription factors in somatic cells enables the induction of induced pluripotent stem cells (iPSCs). This process entails genome-wide remodeling of DNA methylation, chromatin, and transcription. Recent work suggests that the number of active X chromosomes present in a cell influences remodeling of DNA methylation during somatic cell reprogramming to mouse iPSCs. Female iPSCs with 2 active X chromosomes display global DNA hypomethylation, whereas male XY iPSCs show DNA methylation levels similar to the somatic cells they are derived from. Global DNA methylation erasure in female iPSCs takes place genome-wide and involves repression of DNA methyltransferases. However, on loss of one X chromosome, female iPSCs acquire a DNA methylation landscape resembling that of XY iPSCs. Therefore, it is the X chromosome dosage that dictates global DNA methylation levels in iPSCs. Here, we discuss the evidence that links X chromosome dosage with the regulation of DNA methylation in pluripotent stem cells. We focus on iPSCs reprogramming studies, where X chromosome status is a novel factor impacting our understanding of epigenetic remodeling., Competing Interests: Declaration of conflicting interests:The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2018
- Full Text
- View/download PDF
36. Mapping Metabolism: Monitoring Lactate Dehydrogenase Activity Directly in Tissue.
- Author
-
Jelinek D, Flores A, Uebelhoer M, Pasque V, Plath K, Iruela-Arispe ML, Christofk HR, Lowry WE, and Coller HA
- Subjects
- Animals, Mice, L-Lactate Dehydrogenase metabolism
- Abstract
Mapping enzymatic activity in space and time is critical for understanding the molecular basis of cell behavior in normal tissue and disease. In situ metabolic activity assays can provide information about the spatial distribution of metabolic activity within a tissue. We provide here a detailed protocol for monitoring the activity of the enzyme lactate dehydrogenase directly in tissue samples. Lactate dehydrogenase is an important determinant of whether consumed glucose will be converted to energy via aerobic or anaerobic glycolysis. A solution containing lactate and NAD is provided to a frozen tissue section. Cells with high lactate dehydrogenase activity will convert the provided lactate to pyruvate, while simultaneously converting provided nicotinamide adenine dinucleotide (NAD) to NADH and a proton, which can be detected based on the reduction of nitrotetrazolium blue to formazan, which is visualized as a blue precipitate. We describe a detailed protocol for monitoring lactate dehydrogenase activity in mouse skin. Applying this protocol, we found that lactate dehydrogenase activity is high in the quiescent hair follicle stem cells within the skin. Applying the protocol to cultured mouse embryonic stem cells revealed higher staining in cultured embryonic stem cells than mouse embryonic fibroblasts. Analysis of freshly isolated mouse aorta revealed staining in smooth muscle cells perpendicular to the aorta. The methodology provided can be used to spatially map the activity of enzymes that generate a proton in frozen or fresh tissue.
- Published
- 2018
- Full Text
- View/download PDF
37. X Chromosome Dosage Influences DNA Methylation Dynamics during Reprogramming to Mouse iPSCs.
- Author
-
Pasque V, Karnik R, Chronis C, Petrella P, Langerman J, Bonora G, Song J, Vanheer L, Sadhu Dimashkie A, Meissner A, and Plath K
- Subjects
- Animals, Binding Sites, CpG Islands genetics, Embryonic Stem Cells metabolism, Enhancer Elements, Genetic genetics, Female, Genome, Genomic Imprinting, Induced Pluripotent Stem Cells cytology, Male, Mice, Transcription Factors metabolism, Cellular Reprogramming genetics, Chromosomes, Mammalian genetics, DNA Methylation genetics, Induced Pluripotent Stem Cells metabolism, X Chromosome genetics
- Abstract
A dramatic difference in global DNA methylation between male and female cells characterizes mouse embryonic stem cells (ESCs), unlike somatic cells. We analyzed DNA methylation changes during reprogramming of male and female somatic cells and in resulting induced pluripotent stem cells (iPSCs). At an intermediate reprogramming stage, somatic and pluripotency enhancers are targeted for partial methylation and demethylation. Demethylation within pluripotency enhancers often occurs at ESC binding sites of pluripotency transcription factors. Late in reprogramming, global hypomethylation is induced in a female-specific manner. Genome-wide hypomethylation in female cells affects many genomic landmarks, including enhancers and imprint control regions, and accompanies the reactivation of the inactive X chromosome. The loss of one of the two X chromosomes in propagating female iPSCs is associated with genome-wide methylation gain. Collectively, our findings highlight the dynamic regulation of DNA methylation at enhancers during reprogramming and reveal that X chromosome dosage dictates global DNA methylation levels in iPSCs., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
38. Parallel derivation of isogenic human primed and naive induced pluripotent stem cells.
- Author
-
Kilens S, Meistermann D, Moreno D, Chariau C, Gaignerie A, Reignier A, Lelièvre Y, Casanova M, Vallot C, Nedellec S, Flippe L, Firmin J, Song J, Charpentier E, Lammers J, Donnart A, Marec N, Deb W, Bihouée A, Le Caignec C, Pecqueur C, Redon R, Barrière P, Bourdon J, Pasque V, Soumillon M, Mikkelsen TS, Rougeulle C, Fréour T, and David L
- Subjects
- Animals, Blastocyst metabolism, Cells, Cultured, Cellular Reprogramming genetics, Cellular Reprogramming Techniques, Embryonic Development genetics, Embryonic Stem Cells metabolism, Female, Fibroblasts cytology, Fibroblasts metabolism, Germ Layers metabolism, Humans, Induced Pluripotent Stem Cells metabolism, Male, Mice, Transcriptome, Blastocyst cytology, Embryonic Stem Cells cytology, Germ Layers cytology, Induced Pluripotent Stem Cells cytology
- Abstract
Induced pluripotent stem cells (iPSCs) have considerably impacted human developmental biology and regenerative medicine, notably because they circumvent the use of cells of embryonic origin and offer the potential to generate patient-specific pluripotent stem cells. However, conventional reprogramming protocols produce developmentally advanced, or primed, human iPSCs (hiPSCs), restricting their use to post-implantation human development modeling. Hence, there is a need for hiPSCs resembling preimplantation naive epiblast. Here, we develop a method to generate naive hiPSCs directly from somatic cells, using OKMS overexpression and specific culture conditions, further enabling parallel generation of their isogenic primed counterparts. We benchmark naive hiPSCs against human preimplantation epiblast and reveal remarkable concordance in their transcriptome, dependency on mitochondrial respiration and X-chromosome status. Collectively, our results are essential for the understanding of pluripotency regulation throughout preimplantation development and generate new opportunities for disease modeling and regenerative medicine.
- Published
- 2018
- Full Text
- View/download PDF
39. Gene Resistance to Transcriptional Reprogramming following Nuclear Transfer Is Directly Mediated by Multiple Chromatin-Repressive Pathways.
- Author
-
Jullien J, Vodnala M, Pasque V, Oikawa M, Miyamoto K, Allen G, David SA, Brochard V, Wang S, Bradshaw C, Koseki H, Sartorelli V, Beaujean N, and Gurdon J
- Subjects
- 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, Cell Nucleus metabolism, Cellular Reprogramming, Chromatin metabolism, DNA Methylation, Epigenesis, Genetic, Histones metabolism, Nuclear Transfer Techniques, Transcription, Genetic
- 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., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
40. X chromosome reactivation in reprogramming and in development.
- Author
-
Pasque V and Plath K
- Subjects
- Animals, Cell Differentiation genetics, Humans, Models, Biological, Pluripotent Stem Cells, X Chromosome Inactivation, Cellular Reprogramming, X Chromosome
- Abstract
Dramatic epigenetic changes take place during mammalian differentiation from the naïve pluripotent state including the silencing of one of the two X chromosomes in female cells through X chromosome inactivation. Conversely, reprogramming of somatic cells to naive pluripotency is coupled to X chromosome reactivation (XCR). Recent studies in the mouse system have shed light on the mechanisms of XCR by uncovering the timing and steps of XCR during reprogramming to induced pluripotent stem cells (iPSCs), allowing the generation of testable hypotheses during embryogenesis. In contrast, analyses of the X chromosome in human iPSCs have revealed important differences between mouse and human reprogramming processes that can partially be explained by the establishment of distinct pluripotent states and impact disease modeling and the application of human pluripotent stem cells. Here, we review recent literature on XCR as a readout and determinant of reprogramming to pluripotency., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
41. X chromosome reactivation dynamics reveal stages of reprogramming to pluripotency.
- Author
-
Pasque V, Tchieu J, Karnik R, Uyeda M, Sadhu Dimashkie A, Case D, Papp B, Bonora G, Patel S, Ho R, Schmidt R, McKee R, Sado T, Tada T, Meissner A, and Plath K
- Subjects
- Animals, Cdh1 Proteins metabolism, DNA Methylation, Homeodomain Proteins metabolism, Mice, Nanog Homeobox Protein, RNA, Long Noncoding metabolism, Cellular Reprogramming, Epigenesis, Genetic, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, X Chromosome metabolism
- Abstract
Reprogramming to iPSCs resets the epigenome of somatic cells, including the reversal of X chromosome inactivation. We sought to gain insight into the steps underlying the reprogramming process by examining the means by which reprogramming leads to X chromosome reactivation (XCR). Analyzing single cells in situ, we found that hallmarks of the inactive X (Xi) change sequentially, providing a direct readout of reprogramming progression. Several epigenetic changes on the Xi occur in the inverse order of developmental X inactivation, whereas others are uncoupled from this sequence. Among the latter, DNA methylation has an extraordinary long persistence on the Xi during reprogramming, and, like Xist expression, is erased only after pluripotency genes are activated. Mechanistically, XCR requires both DNA demethylation and Xist silencing, ensuring that only cells undergoing faithful reprogramming initiate XCR. Our study defines the epigenetic state of multiple sequential reprogramming intermediates and establishes a paradigm for studying cell fate transitions during reprogramming., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
42. Mitosis gives a brief window of opportunity for a change in gene transcription.
- Author
-
Halley-Stott RP, Jullien J, Pasque V, and Gurdon J
- Subjects
- Amphibians, Animals, Cell Line, Histones metabolism, Mice, Nuclear Transfer Techniques, Oocytes metabolism, Cellular Reprogramming, Chromatin metabolism, Mitosis physiology, Transcription, Genetic
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2014
- Full Text
- View/download PDF
43. A new route to human embryonic stem cells.
- Author
-
Trounson A, Daley GQ, Pasque V, and Plath K
- Subjects
- Cell Differentiation physiology, Embryo Research, Embryonic Stem Cells physiology, Humans, Nuclear Transfer Techniques, Oocytes cytology, Oocytes physiology, Pluripotent Stem Cells physiology, Regenerative Medicine trends, Cellular Reprogramming, Embryonic Stem Cells cytology, Pluripotent Stem Cells cytology, Regenerative Medicine methods
- Published
- 2013
- Full Text
- View/download PDF
44. Nuclear reprogramming.
- Author
-
Halley-Stott RP, Pasque V, and Gurdon JB
- Subjects
- Animals, Cell Membrane metabolism, Cell Transdifferentiation, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Epigenesis, Genetic, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Meiotic Prophase I, Metaphase, Ovum cytology, Transcription, Genetic, Cellular Reprogramming, Nuclear Transfer Techniques, Ovum metabolism
- Abstract
There is currently particular interest in the field of nuclear reprogramming, a process by which the identity of specialised cells may be changed, typically to an embryonic-like state. Reprogramming procedures provide insight into many mechanisms of fundamental cell biology and have several promising applications, most notably in healthcare through the development of human disease models and patient-specific tissue-replacement therapies. Here, we introduce the field of nuclear reprogramming and briefly discuss six of the procedures by which reprogramming may be experimentally performed: nuclear transfer to eggs or oocytes, cell fusion, extract treatment, direct reprogramming to pluripotency and transdifferentiation.
- Published
- 2013
- Full Text
- View/download PDF
45. Histone variant macroH2A marks embryonic differentiation in vivo and acts as an epigenetic barrier to induced pluripotency.
- Author
-
Pasque V, Radzisheuskaya A, Gillich A, Halley-Stott RP, Panamarova M, Zernicka-Goetz M, Surani MA, and Silva JC
- Subjects
- Animals, Cell Differentiation genetics, Cellular Reprogramming, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Epigenomics, Female, Gene Expression Regulation, Developmental, Histones genetics, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism, Transfection, Embryonic Stem Cells physiology, Histones metabolism, Pluripotent Stem Cells physiology
- Abstract
How cell fate becomes restricted during somatic cell differentiation is a long-lasting question in biology. Epigenetic mechanisms not present in pluripotent cells and acquired during embryonic development are expected to stabilize the differentiated state of somatic cells and thereby restrict their ability to convert to another fate. The histone variant macroH2A acts as a component of an epigenetic multilayer that heritably maintains the silent X chromosome and has been shown to restrict tumor development. Here we show that macroH2A marks the differentiated cell state during mouse embryogenesis. MacroH2A.1 was found to be present at low levels upon the establishment of pluripotency in the inner cell mass and epiblast, but it was highly enriched in the trophectoderm and differentiated somatic cells later in mouse development. Chromatin immunoprecipitation revealed that macroH2A.1 is incorporated in the chromatin of regulatory regions of pluripotency genes in somatic cells such as mouse embryonic fibroblasts and adult neural stem cells, but not in embryonic stem cells. Removal of macroH2A.1, macroH2A.2 or both increased the efficiency of induced pluripotency up to 25-fold. The obtained induced pluripotent stem cells reactivated pluripotency genes, silenced retroviral transgenes and contributed to chimeras. In addition, overexpression of macroH2A isoforms prevented efficient reprogramming of epiblast stem cells to naïve pluripotency. In summary, our study identifies for the first time a link between an epigenetic mark and cell fate restriction during somatic cell differentiation, which helps to maintain cell identity and antagonizes induction of a pluripotent stem cell state.
- Published
- 2012
- Full Text
- View/download PDF
46. Epiblast stem cell-based system reveals reprogramming synergy of germline factors.
- Author
-
Gillich A, Bao S, Grabole N, Hayashi K, Trotter MW, Pasque V, Magnúsdóttir E, and Surani MA
- Subjects
- Animals, DNA Methylation, DNA-Binding Proteins, Germ Cells cytology, Germ Layers cytology, Humans, Mice, RNA-Binding Proteins, Stem Cells cytology, Cell Dedifferentiation, Epigenesis, Genetic, Germ Layers metabolism, Kruppel-Like Transcription Factors metabolism, Repressor Proteins metabolism, Stem Cells metabolism, Transcription Factors metabolism
- Abstract
Epigenetic reprogramming in early germ cells is critical toward the establishment of totipotency, but investigations of the germline events are intractable. An objective cell culture-based system could provide mechanistic insight on how the key determinants of primordial germ cells (PGCs), including Prdm14, induce reprogramming in germ cells to an epigenetic ground state. Here we show a Prdm14-Klf2 synergistic effect that can accelerate and enhance reversion of mouse epiblast stem cells (epiSCs) to a naive pluripotent state, including X reactivation and DNA demethylation. Notably, Prdm14 alone has little effect on epiSC reversion, but it enhances the competence for reprogramming and potentially PGC specification. Reprogramming of epiSCs by the combinatorial effect of Prdm14-Klf2 involves key epigenetic changes, which might have an analogous role in PGCs. Our study provides a paradigm toward a systematic analysis of how other key genes contribute to complex and dynamic events of reprogramming in the germline., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
47. RUNX3, EGR1 and SOX9B form a regulatory cascade required to modulate BMP-signaling during cranial cartilage development in zebrafish.
- Author
-
Dalcq J, Pasque V, Ghaye A, Larbuisson A, Motte P, Martial JA, and Muller M
- Subjects
- Alcian Blue pharmacology, Animals, Cartilage metabolism, Cell Differentiation, Endoderm metabolism, Epithelium metabolism, Female, Follistatin metabolism, Male, Neural Crest cytology, Oligonucleotides metabolism, Pyrazoles metabolism, Pyrimidines metabolism, Signal Transduction, Skull embryology, Skull metabolism, Time Factors, Zebrafish, Bone Morphogenetic Proteins metabolism, Core Binding Factor Alpha 3 Subunit metabolism, Early Growth Response Protein 1 metabolism, Gene Expression Regulation, Developmental, SOX9 Transcription Factor metabolism, Zebrafish Proteins metabolism
- Abstract
The cartilaginous elements forming the pharyngeal arches of the zebrafish derive from cranial neural crest cells. Their proper differentiation and patterning are regulated by reciprocal interactions between neural crest cells and surrounding endodermal, ectodermal and mesodermal tissues. In this study, we show that the endodermal factors Runx3 and Sox9b form a regulatory cascade with Egr1 resulting in transcriptional repression of the fsta gene, encoding a BMP antagonist, in pharyngeal endoderm. Using a transgenic line expressing a dominant negative BMP receptor or a specific BMP inhibitor (dorsomorphin), we show that BMP signaling is indeed required around 30 hpf in the neural crest cells to allow cell differentiation and proper pharyngeal cartilage formation. Runx3, Egr1, Sox9b and BMP signaling are required for expression of runx2b, one of the key regulator of cranial cartilage maturation and bone formation. Finally, we show that egr1 depletion leads to increased expression of fsta and inhibition of BMP signaling in the pharyngeal region. In conclusion, we show that the successive induction of the transcription factors Runx3, Egr1 and Sox9b constitutes a regulatory cascade that controls expression of Follistatin A in pharyngeal endoderm, the latter modulating BMP signaling in developing cranial cartilage in zebrafish.
- Published
- 2012
- Full Text
- View/download PDF
48. Epigenetic factors influencing resistance to nuclear reprogramming.
- Author
-
Pasque V, Jullien J, Miyamoto K, Halley-Stott RP, and Gurdon JB
- Subjects
- Animals, Cell Division, DNA Methylation, Humans, Models, Genetic, Transcription, Genetic, Cellular Reprogramming, Epigenesis, Genetic
- Abstract
Patient-specific somatic cell reprogramming is likely to have a large impact on medicine by providing a source of cells for disease modelling and regenerative medicine. Several strategies can be used to reprogram cells, yet they are generally characterised by a low reprogramming efficiency, reflecting the remarkable stability of the differentiated state. Transcription factors, chromatin modifications, and noncoding RNAs can increase the efficiency of reprogramming. However, the success of nuclear reprogramming is limited by epigenetic mechanisms that stabilise the state of gene expression in somatic cells and thereby resist efficient reprogramming. We review here the factors that influence reprogramming efficiency, especially those that restrict the natural reprogramming mechanisms of eggs and oocytes. We see this as a step towards understanding the mechanisms by which nuclear reprogramming takes place., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
49. Epigenetic stability of repressed states involving the histone variant macroH2A revealed by nuclear transfer to Xenopus oocytes.
- Author
-
Pasque V, Halley-Stott RP, Gillich A, Garrett N, and Gurdon JB
- Subjects
- Animals, Embryo, Nonmammalian cytology, Embryo, Nonmammalian metabolism, Histones genetics, Oocytes cytology, Repressor Proteins genetics, Transcription, Genetic physiology, X Chromosome genetics, X Chromosome metabolism, Xenopus Proteins genetics, Xenopus laevis, Epigenesis, Genetic physiology, Histones metabolism, Nuclear Transfer Techniques, Oocytes metabolism, Repressor Proteins metabolism, Xenopus Proteins metabolism
- Abstract
How various epigenetic mechanisms restrict chromatin plasticity to determine the stability of repressed genes is poorly understood. Nuclear transfer to Xenopus oocytes induces the transcriptional reactivation of previously silenced genes. Recent work suggests that it can be used to analyze the epigenetic stability of repressed states. The notion that the epigenetic state of genes is an important determinant of the efficiency of nuclear reprogramming is supported by the differential reprogramming of given genes from different starting epigenetic configurations. After nuclear transfer, transcription from the inactive X chromosome of post-implantation-derived epiblast stem cells is reactivated. However, the same chromosome is resistant to reactivation when embryonic fibroblasts are used. Here, we discuss different kinds of evidence that link the histone variant macroH2A to the increased stability of repressed states. We focus on developmentally regulated X chromosome inactivation and repression of autosomal pluripotency genes, where macroH2A may help maintain the long-term stability of the differentiated state of somatic cells.
- Published
- 2011
- Full Text
- View/download PDF
50. Epigenetic reprogramming: is deamination key to active DNA demethylation?
- Author
-
Teperek-Tkacz M, Pasque V, Gentsch G, and Ferguson-Smith AC
- Subjects
- APOBEC-1 Deaminase, Animals, Cytidine Deaminase genetics, Cytidine Deaminase metabolism, Cytidine Deaminase physiology, DNA Methylation genetics, Deamination genetics, Humans, Models, Biological, Vertebrates genetics, Vertebrates metabolism, Vertebrates physiology, AICDA (Activation-Induced Cytidine Deaminase), Cellular Reprogramming physiology, DNA Methylation physiology, Deamination physiology, Epigenesis, Genetic physiology
- Abstract
DNA demethylation processes are important for reproduction, being central in epigenetic reprogramming during embryonic and germ cell development. While the enzymes methylating DNA have been known for many years, identification of factors capable of mediating active DNA demethylation has been challenging. Recent findings suggest that cytidine deaminases may be key players in active DNA demethylation. One of the most investigated candidates is activation-induced cytidine deaminase (AID), best known for its role in generating secondary antibody diversity in B cells. We evaluate evidence for cytidine deaminases in DNA demethylation pathways in vertebrates and discuss possible models for their targeting and activity regulation. These findings are also considered along with alternative demethylation pathways involving hydroxymethylation.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.