Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 3550, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729116

ABSTRACT

The epiblast is the source of all mammalian embryonic tissues and of pluripotent embryonic stem cells. It differentiates alongside the primitive endoderm in a "salt and pepper" pattern from inner cell mass (ICM) progenitors during the preimplantation stages through the activity of NANOG, GATA6 and the FGF pathway. When and how epiblast lineage specification is initiated is still unclear. Here, we show that the coordinated expression of pluripotency markers defines epiblast identity. Conversely, ICM progenitor cells display random cell-to-cell variability in expression of various pluripotency markers, remarkably dissimilar from the epiblast signature and independently from NANOG, GATA6 and FGF activities. Coordination of pluripotency markers expression fails in Nanog and Gata6 double KO (DKO) embryos. Collectively, our data suggest that NANOG triggers epiblast specification by ensuring the coordinated expression of pluripotency markers in a subset of cells, implying a stochastic mechanism. These features are likely conserved, as suggested by analysis of human embryos.


Subject(s)
Endoderm , Germ Layers , Animals , Blastocyst/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Endoderm/metabolism , Gene Expression Regulation, Developmental , Germ Layers/metabolism , Humans , Mammals/genetics , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism
2.
Cell Stem Cell ; 28(9): 1625-1640.e6, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34004179

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.


Subject(s)
Embryonic Development , Transcriptome , Animals , Blastocyst , Cell Lineage/genetics , Embryonic Development/genetics , Germ Layers , Humans , Mice , Transcriptome/genetics
3.
Cell Rep ; 33(8): 108419, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33238118

ABSTRACT

Human trophoblast stem cells (hTSCs) derived from blastocysts and first-trimester cytotrophoblasts offer an unprecedented opportunity to study the placenta. However, access to human embryos and first-trimester placentas is limited, thus preventing the establishment of hTSCs from diverse genetic backgrounds associated with placental disorders. Here, we show that hTSCs can be generated from numerous genetic backgrounds using post-natal cells via two alternative methods: (1) somatic cell reprogramming of adult fibroblasts with OCT4, SOX2, KLF4, MYC (OSKM) and (2) cell fate conversion of naive and extended pluripotent stem cells. The resulting induced/converted hTSCs recapitulated hallmarks of hTSCs including long-term self-renewal, expression of specific transcription factors, transcriptomic signature, and the potential to differentiate into syncytiotrophoblast and extravillous trophoblast cells. We also clarified the developmental stage of hTSCs and show that these cells resemble day 8 cytotrophoblasts. Altogether, hTSC lines of diverse genetic origins open the possibility to model both placental development and diseases in a dish.


Subject(s)
Pluripotent Stem Cells/metabolism , Trophoblasts/metabolism , Cell Differentiation , Female , Humans , Pregnancy
4.
Nutrients ; 12(5)2020 May 19.
Article in English | MEDLINE | ID: mdl-32438566

ABSTRACT

Fetal brain development is closely dependent on maternal nutrition and metabolic status. Maternal protein restriction (PR) is known to be associated with alterations in the structure and function of the hypothalamus, leading to impaired control of energy homeostasis and food intake. The objective of this study was to identify the cellular and molecular systems underlying these effects during fetal development. We combined a global transcriptomic analysis on the fetal hypothalamus from a rat model of maternal PR with in vitro neurosphere culture and cellular analyses. Several genes encoding proteins from the mitochondrial respiratory chain complexes were overexpressed in the PR group and mitochondrial metabolic activity in the fetal hypothalamus was altered. The level of the N6-methyladenosine epitranscriptomic mark was reduced in the PR fetuses, and the expression of several genes involved in the writing/erasing/reading of this mark was indeed altered, as well as genes encoding several RNA-binding proteins. Additionally, we observed a higher number of neuronal-committed progenitors at embryonic day 17 (E17) in the PR fetuses. Together, these data strongly suggest a metabolic adaptation to the amino acid shortage, combined with the post-transcriptional control of protein expression, which might reflect alterations in the control of the timing of neuronal progenitor differentiation.


Subject(s)
Diet, Protein-Restricted/adverse effects , Fetus/metabolism , Hypothalamus/embryology , Maternal Nutritional Physiological Phenomena/genetics , Mitochondria/genetics , Animals , Female , Fetal Development/genetics , Hypothalamus/metabolism , Pregnancy , Rats
5.
Nat Commun ; 9(1): 360, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367672

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.


Subject(s)
Blastocyst/cytology , Embryonic Stem Cells/cytology , Germ Layers/cytology , Induced Pluripotent Stem Cells/cytology , 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
6.
JCI Insight ; 2(3): e90088, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28194440

ABSTRACT

Rat and human CD4+ and CD8+ Tregs expressing low levels of CD45RC have strong immunoregulatory properties. We describe here that human CD45 isoforms are nonredundant and identify distinct subsets of cells. We show that CD45RC is not expressed by CD4+ and CD8+ Foxp3+ Tregs, while CD45RA/RB/RO are. Transient administration of a monoclonal antibody (mAb) targeting CD45RC in a rat cardiac allotransplantation model induced transplant tolerance associated with inhibition of allogeneic humoral responses but maintained primary and memory responses against cognate antigens. Anti-CD45RC mAb induced rapid death of CD45RChigh T cells through intrinsic cell signaling but preserved and potentiated CD4+ and CD8+ CD45RClow/- Tregs, which are able to adoptively transfer donor-specific tolerance to grafted recipients. Anti-CD45RC treatment results in distinct transcriptional signature of CD4+ and CD8+ CD45RClow/- Tregs. Finally, we demonstrate that anti-human CD45RC treatment inhibited graft-versus-host disease (GVHD) in immune-humanized NSG mice. Thus, short-term anti-CD45RC is a potent therapeutic candidate to induce transplantation tolerance in human.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Graft vs Host Disease/drug therapy , Leukocyte Common Antigens/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Heart Transplantation , Humans , Immunity, Humoral/drug effects , Mice , Rats , Transplantation Tolerance
7.
Front Immunol ; 8: 2014, 2017.
Article in English | MEDLINE | ID: mdl-29445370

ABSTRACT

Both CD4+ and CD8+ Tregs play a critical role in the control of immune responses and immune tolerance; however, our understanding of CD8+ Tregs is limited while they are particularly promising for therapeutic application. We report here existence of highly suppressive human CD8+CD45RClow/- Tregs expressing Foxp3 and producing IFNγ, IL-10, IL-34, and TGFß to mediate their suppressive activity. We demonstrate that total CD8+CD45RClow/- Tregs can be efficiently expanded in the presence of anti-CD3/28 mAbs, high-dose IL-2 and IL-15 and that such expanded Tregs efficiently delay GVHD and human skin transplantation rejection in immune humanized mice. Robustly expanded CD8+ Tregs displayed a specific gene signature, upregulated cytokines and expansion in the presence of rapamycin greatly improved proliferation and suppression. We show that CD8+CD45RClow/- Tregs are equivalent to canonical CD4+CD25highCD127low/- Tregs for suppression of allogeneic immune responses in vitro. Altogether, our results open new perspectives to tolerogenic strategies in human solid organ transplantation and GVHD.

SELECTION OF CITATIONS
SEARCH DETAIL
...