Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 185(5): 777-793.e20, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35196500

ABSTRACT

In development, lineage segregation is coordinated in time and space. An important example is the mammalian inner cell mass, in which the primitive endoderm (PrE, founder of the yolk sac) physically segregates from the epiblast (EPI, founder of the fetus). While the molecular requirements have been well studied, the physical mechanisms determining spatial segregation between EPI and PrE remain elusive. Here, we investigate the mechanical basis of EPI and PrE sorting. We find that rather than the differences in static cell surface mechanical parameters as in classical sorting models, it is the differences in surface fluctuations that robustly ensure physical lineage sorting. These differential surface fluctuations systematically correlate with differential cellular fluidity, which we propose together constitute a non-equilibrium sorting mechanism for EPI and PrE lineages. By combining experiments and modeling, we identify cell surface dynamics as a key factor orchestrating the correct spatial segregation of the founder embryonic lineages.


Subject(s)
Blastocyst , Embryo, Mammalian , Endoderm , Animals , Blastocyst/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Membrane/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Endoderm/metabolism , Mammals , Mice , Protein Transport
3.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33452132

ABSTRACT

OCT4 is a fundamental component of the molecular circuitry governing pluripotency in vivo and in vitro. To determine how OCT4 establishes and protects the pluripotent lineage in the embryo, we used comparative single-cell transcriptomics and quantitative immunofluorescence on control and OCT4 null blastocyst inner cell masses at two developmental stages. Surprisingly, activation of most pluripotency-associated transcription factors in the early mouse embryo occurs independently of OCT4, with the exception of the JAK/STAT signaling machinery. Concurrently, OCT4 null inner cell masses ectopically activate a subset of trophectoderm-associated genes. Inspection of metabolic pathways implicates the regulation of rate-limiting glycolytic enzymes by OCT4, consistent with a role in sustaining glycolysis. Furthermore, up-regulation of the lysosomal pathway was specifically detected in OCT4 null embryos. This finding implicates a requirement for OCT4 in the production of normal trophectoderm. Collectively, our findings uncover regulation of cellular metabolism and biophysical properties as mechanisms by which OCT4 instructs pluripotency.


Subject(s)
Cell Lineage/genetics , Embryonic Development/immunology , Octamer Transcription Factor-3/genetics , STAT3 Transcription Factor/genetics , Animals , Blastocyst Inner Cell Mass/metabolism , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Glycolysis/genetics , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics , Single-Cell Analysis
4.
Development ; 146(7)2019 04 03.
Article in English | MEDLINE | ID: mdl-30944104

ABSTRACT

Human naïve pluripotent stem cells (PSCs) share features with the pre-implantation epiblast. They therefore provide an unmatched opportunity for characterising the developmental programme of pluripotency in Homo sapiens Here, we confirm that naïve PSCs do not respond directly to germ layer induction, but must first acquire competence. Capacitation for multi-lineage differentiation occurs without exogenous growth factor stimulation and is facilitated by inhibition of Wnt signalling. Whole-transcriptome profiling during this formative transition highlights dynamic changes in gene expression, which affect many cellular properties including metabolism and epithelial features. Notably, naïve pluripotency factors are exchanged for postimplantation factors, but competent cells remain devoid of lineage-specific transcription. The gradual pace of transition for human naïve PSCs is consistent with the timespan of primate development from blastocyst to gastrulation. Transcriptome trajectory during in vitro capacitation of human naïve cells tracks the progression of the epiblast during embryogenesis in Macaca fascicularis, but shows greater divergence from mouse development. Thus, the formative transition of naïve PSCs in a simple culture system may recapitulate essential and specific features of pluripotency dynamics during an inaccessible period of human embryogenesis.


Subject(s)
Pluripotent Stem Cells/cytology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation/physiology , Cell Lineage , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/metabolism , Humans , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/physiology
5.
Development ; 145(3)2018 02 07.
Article in English | MEDLINE | ID: mdl-29361568

ABSTRACT

Single-cell profiling techniques create opportunities to delineate cell fate progression in mammalian development. Recent studies have provided transcriptome data from human pre-implantation embryos, in total comprising nearly 2000 individual cells. Interpretation of these data is confounded by biological factors, such as variable embryo staging and cell-type ambiguity, as well as technical challenges in the collective analysis of datasets produced with different sample preparation and sequencing protocols. Here, we address these issues to assemble a complete gene expression time course spanning human pre-implantation embryogenesis. We identify key transcriptional features over developmental time and elucidate lineage-specific regulatory networks. We resolve post-hoc cell-type assignment in the blastocyst, and define robust transcriptional prototypes that capture epiblast and primitive endoderm lineages. Examination of human pluripotent stem cell transcriptomes in this framework identifies culture conditions that sustain a naïve state pertaining to the inner cell mass. Our approach thus clarifies understanding both of lineage segregation in the early human embryo and of in vitro stem cell identity, and provides an analytical resource for comparative molecular embryology.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cell Line , Cell Lineage/genetics , Cell Lineage/physiology , Chromosome Mapping , Embryo Culture Techniques , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Profiling , Genetic Markers , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Primates , Single-Cell Analysis
6.
Development ; 145(21)2018 11 09.
Article in English | MEDLINE | ID: mdl-30413530

ABSTRACT

The mouse embryo is the canonical model for mammalian preimplantation development. Recent advances in single cell profiling allow detailed analysis of embryogenesis in other eutherian species, including human, to distinguish conserved from divergent regulatory programs and signalling pathways in the rodent paradigm. Here, we identify and compare transcriptional features of human, marmoset and mouse embryos by single cell RNA-seq. Zygotic genome activation correlates with the presence of polycomb repressive complexes in all three species, while ribosome biogenesis emerges as a predominant attribute in primate embryos, supporting prolonged translation of maternally deposited RNAs. We find that transposable element expression signatures are species, stage and lineage specific. The pluripotency network in the primate epiblast lacks certain regulators that are operative in mouse, but encompasses WNT components and genes associated with trophoblast specification. Sequential activation of GATA6, SOX17 and GATA4 markers of primitive endoderm identity is conserved in primates. Unexpectedly, OTX2 is also associated with primitive endoderm specification in human and non-human primate blastocysts. Our cross-species analysis demarcates both conserved and primate-specific features of preimplantation development, and underscores the molecular adaptability of early mammalian embryogenesis.


Subject(s)
Callithrix/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Single-Cell Analysis , Transcriptome/genetics , Animals , Conserved Sequence/genetics , Endoderm/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Germ Layers/metabolism , Humans , Mice , Otx Transcription Factors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics , Transcription, Genetic
7.
Am J Physiol Heart Circ Physiol ; 310(11): H1622-48, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26993221

ABSTRACT

c-Kit(pos) cardiac progenitor cells (CPCs) represent a successful approach in healing the infarcted heart and rescuing its mechanical function, but electrophysiological consequences are uncertain. CPC mobilization promoted by hepatocyte growth factor (HGF) and IGF-1 improved electrogenesis in myocardial infarction (MI). We hypothesized that locally delivered CPCs supplemented with HGF + IGF-1 (GFs) can concur in ameliorating electrical stability of the regenerated heart. Adult male Wistar rats (139 rats) with 4-wk-old MI or sham conditions were randomized to receive intramyocardial injection of GFs, CPCs, CPCs + GFs, or vehicle (V). Enhanced green fluorescent protein-tagged CPCs were used for cell tracking. Vulnerability to stress-induced arrhythmia was assessed by telemetry-ECG. Basic cardiac electrophysiological properties were examined by epicardial multiple-lead recording. Hemodynamic function was measured invasively. Hearts were subjected to anatomical, morphometric, immunohistochemical, and molecular biology analyses. Compared with V and at variance with individual CPCs, CPCs + GFs approximately halved arrhythmias in all animals, restoring cardiac anisotropy toward sham values. GFs alone reduced arrhythmias by less than CPCs + GFs, prolonging ventricular refractoriness without affecting conduction velocity. Concomitantly, CPCs + GFs reactivated the expression levels of Connexin-43 and Connexin-40 as well as channel proteins of key depolarizing and repolarizing ion currents differently than sole GFs. Mechanical function and anatomical remodeling were equally improved by all regenerative treatments, thus exhibiting a divergent behavior relative to electrical aspects. Conclusively, we provided evidence of distinctive antiarrhythmic action of locally injected GF-supplemented CPCs, likely attributable to retrieval of Connexin-43, Connexin-40, and Cav1.2 expression, favoring intercellular coupling and spread of excitation in mended heart.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Hepatocyte Growth Factor/therapeutic use , Insulin-Like Growth Factor I/therapeutic use , Myocardial Infarction/therapy , Stem Cells , Animals , Connexin 43/metabolism , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar
9.
iScience ; 24(3): 102153, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33665571

ABSTRACT

Induced pluripotency provides a tool to explore mechanisms underlying establishment, maintenance, and differentiation of naive pluripotent stem cells (nPSCs). Here, we report that self-renewal of nPSCs requires minimal Sox2 expression (Sox2-low). Sox2-low nPSCs do not show impaired neuroectoderm specification and differentiate efficiently in vitro into all embryonic germ lineages. Strikingly, upon the removal of self-renewing cues Sox2-low nPSCs differentiate into both embryonic and extraembryonic cell fates in vitro and in vivo. This differs from previous studies which only identified conditions that allowed cells to differentiate to one fate or the other. At the single-cell level self-renewing Sox2-low nPSCs exhibit a naive molecular signature. However, they display a nearer trophoblast identity than controls and decreased ability of Oct4 to bind naïve-associated regulatory sequences. In sum, this work defines wild-type levels of Sox2 as a restrictor of developmental potential and suggests perturbation of naive network as a mechanism to increase cell plasticity.

10.
Nat Commun ; 12(1): 6132, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675200

ABSTRACT

Studies of mechanical signalling are typically performed by comparing cells cultured on soft and stiff hydrogel-based substrates. However, it is challenging to independently and robustly control both substrate stiffness and extracellular matrix tethering to substrates, making matrix tethering a potentially confounding variable in mechanical signalling investigations. Moreover, unstable matrix tethering can lead to poor cell attachment and weak engagement of cell adhesions. To address this, we developed StemBond hydrogels, a hydrogel in which matrix tethering is robust and can be varied independently of stiffness. We validate StemBond hydrogels by showing that they provide an optimal system for culturing mouse and human pluripotent stem cells. We further show how soft StemBond hydrogels modulate stem cell function, partly through stiffness-sensitive ERK signalling. Our findings underline how substrate mechanics impact mechanosensitive signalling pathways regulating self-renewal and differentiation, indicating that optimising the complete mechanical microenvironment will offer greater control over stem cell fate specification.


Subject(s)
Cell Culture Techniques/instrumentation , Extracellular Matrix/chemistry , Hydrogels/chemistry , Pluripotent Stem Cells/cytology , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Mechanotransduction, Cellular , Mice , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/metabolism
11.
Cell Stem Cell ; 25(3): 388-406.e8, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31422912

ABSTRACT

Understanding how cell identity transitions occur and whether there are multiple paths between the same beginning and end states are questions of wide interest. Here we show that acquisition of naive pluripotency can follow transcriptionally and mechanistically distinct routes. Starting from post-implantation epiblast stem cells (EpiSCs), one route advances through a mesodermal state prior to naive pluripotency induction, whereas another transiently resembles the early inner cell mass and correspondingly gains greater developmental potency. These routes utilize distinct signaling networks and transcription factors but subsequently converge on the same naive endpoint, showing surprising flexibility in mechanisms underlying identity transitions and suggesting that naive pluripotency is a multidimensional attractor state. These route differences are reconciled by precise expression of Oct4 as a unifying, essential, and sufficient feature. We propose that fine-tuned regulation of this "transition factor" underpins multidimensional access to naive pluripotency, offering a conceptual framework for understanding cell identity transitions.


Subject(s)
Blastocyst Inner Cell Mass/physiology , Germ Layers/physiology , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation , Cell Line , Cell Plasticity , Cellular Reprogramming , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL