Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
Add more filters

Publication year range
1.
Cell ; 165(4): 1012-26, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27062923

ABSTRACT

Mouse studies have been instrumental in forming our current understanding of early cell-lineage decisions; however, similar insights into the early human development are severely limited. Here, we present a comprehensive transcriptional map of human embryo development, including the sequenced transcriptomes of 1,529 individual cells from 88 human preimplantation embryos. These data show that cells undergo an intermediate state of co-expression of lineage-specific genes, followed by a concurrent establishment of the trophectoderm, epiblast, and primitive endoderm lineages, which coincide with blastocyst formation. Female cells of all three lineages achieve dosage compensation of X chromosome RNA levels prior to implantation. However, in contrast to the mouse, XIST is transcribed from both alleles throughout the progression of this expression dampening, and X chromosome genes maintain biallelic expression while dosage compensation proceeds. We envision broad utility of this transcriptional atlas in future studies on human development as well as in stem cell research.


Subject(s)
Blastocyst/metabolism , Chromosomes, Human, X , Single-Cell Analysis , Blastocyst Inner Cell Mass/metabolism , Dosage Compensation, Genetic , Female , Humans , Male , RNA, Long Noncoding/genetics , Sequence Analysis, RNA , Sex Characteristics , Transcriptome
2.
Cell ; 165(6): 1375-1388, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259149

ABSTRACT

How the chromatin regulatory landscape in the inner cell mass cells is established from differentially packaged sperm and egg genomes during preimplantation development is unknown. Here, we develop a low-input DNase I sequencing (liDNase-seq) method that allows us to generate maps of DNase I-hypersensitive site (DHS) of mouse preimplantation embryos from 1-cell to morula stage. The DHS landscape is progressively established with a drastic increase at the 8-cell stage. Paternal chromatin accessibility is quickly reprogrammed after fertilization to the level similar to maternal chromatin, while imprinted genes exhibit allelic accessibility bias. We demonstrate that transcription factor Nfya contributes to zygotic genome activation and DHS formation at the 2-cell stage and that Oct4 contributes to the DHSs gained at the 8-cell stage. Our study reveals the dynamic chromatin regulatory landscape during early development and identifies key transcription factors important for DHS establishment in mammalian embryos.


Subject(s)
Blastocyst , Chromatin/metabolism , Animals , Binding Sites , Blastocyst/cytology , Blastocyst Inner Cell Mass/metabolism , CCAAT-Binding Factor/metabolism , Chromosome Mapping , DNA/metabolism , Deoxyribonuclease I/metabolism , Embryonic Development , Female , Gene Expression Regulation, Developmental , Male , Mice , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic
3.
Nature ; 587(7834): 443-447, 2020 11.
Article in English | MEDLINE | ID: mdl-32968278

ABSTRACT

Current understandings of cell specification in early mammalian pre-implantation development are based mainly on mouse studies. The first lineage differentiation event occurs at the morula stage, with outer cells initiating a trophectoderm (TE) placental progenitor program. The inner cell mass arises from inner cells during subsequent developmental stages and comprises precursor cells of the embryo proper and yolk sac1. Recent gene-expression analyses suggest that the mechanisms that regulate early lineage specification in the mouse may differ in other mammals, including human2-5 and cow6. Here we show the evolutionary conservation of a molecular cascade that initiates TE segregation in human, cow and mouse embryos. At the morula stage, outer cells acquire an apical-basal cell polarity, with expression of atypical protein kinase C (aPKC) at the contact-free domain, nuclear expression of Hippo signalling pathway effectors and restricted expression of TE-associated factors such as GATA3, which suggests initiation of a TE program. Furthermore, we demonstrate that inhibition of aPKC by small-molecule pharmacological modulation or Trim-Away protein depletion impairs TE initiation at the morula stage. Our comparative embryology analysis provides insights into early lineage specification and suggests that a similar mechanism initiates a TE program in human, cow and mouse embryos.


Subject(s)
Biological Evolution , Ectoderm/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Transcription, Genetic , Trophoblasts/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cattle , Cell Lineage , Cell Polarity , Ectoderm/cytology , Embryo, Mammalian/enzymology , Female , GATA3 Transcription Factor/metabolism , Hippo Signaling Pathway , Humans , Mice , Morula/cytology , Morula/enzymology , Morula/metabolism , Placenta/cytology , Placenta/metabolism , Pregnancy , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transcription Factors/metabolism , Trophoblasts/cytology , YAP-Signaling Proteins , Yolk Sac/cytology , Yolk Sac/metabolism
4.
Hum Reprod ; 39(9): 1889-1898, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38926157

ABSTRACT

In the first days of life, cells of the mammalian embryo segregate into two distinct lineages, trophectoderm and inner cell mass. Unlike nonmammalian species, mammalian development does not proceed from predetermined factors in the oocyte. Rather, asymmetries arise de novo in the early embryo incorporating cues from cell position, contractility, polarity, and cell-cell contacts. Molecular heterogeneities, including transcripts and non-coding RNAs, have now been characterized as early as the 2-cell stage. However, it's debated whether these early heterogeneities bias cells toward one fate or the other or whether lineage identity arises stochastically at the 16-cell stage. This review summarizes what is known about early blastomere asymmetries and our understanding of lineage allocation in the context of historical models. Preimplantation development is reviewed coupled with what is known about changes in morphology, contractility, and transcription factor networks. The addition of single-cell atlases of human embryos has begun to reveal key differences between human and mouse, including the timing of events and core transcription factors. Furthermore, the recent generation of blastoid models will provide valuable tools to test and understand fate determinants. Lastly, new techniques are reviewed, which may better synthesize existing knowledge with emerging data sets and reconcile models with the regulative capacity unique to the mammalian embryo.


Subject(s)
Blastocyst , Cell Lineage , Embryonic Development , Animals , Humans , Blastocyst/cytology , Blastocyst/physiology , Embryonic Development/physiology , Mice , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/physiology , Blastocyst Inner Cell Mass/metabolism , Gene Expression Regulation, Developmental , Blastomeres/cytology , Blastomeres/physiology , Blastomeres/metabolism , Mammals , Embryo, Mammalian/cytology
5.
Reproduction ; 167(6)2024 06 01.
Article in English | MEDLINE | ID: mdl-38552319

ABSTRACT

In brief: MEK signalling pathway is required for hypoblast differentiation in mouse embryos, but its role in ungulate embryos remains controversial. This paper demonstrates that MEK is required for hypoblast specification in the inner cell mass of the ovine blastocyst and that it plays a role during the hypoblast migration occurring following blastocyst hatching. Abstract: Early embryo development requires the differentiation of three cell lineages in two differentiation events. The second lineage specification differentiates the inner cell mass into epiblast, which will form the proper fetus, and hypoblast, which together with the trophectoderm will form the extraembryonic membranes and the fetal part of the placenta. MEK signalling pathway is required for hypoblast differentiation in mouse embryos, but its role in ungulate embryos remains controversial. The aim of this work was to analyse the role of MEK signalling on hypoblast specification at the blastocyst stage and on hypoblast migration during post-hatching stages in vitro in the ovine species. Using well-characterized and reliable lineage markers, and different MEK inhibitor concentrations, we demonstrate that MEK signalling pathway is required for hypoblast specification in the inner cell mass of the ovine blastocyst, and that it plays a role during the hypoblast migration occurring following blastocyst hatching. These results show that the role of MEK signalling pathway on hypoblast specification is conserved in phylogenetically distant mammals.


Subject(s)
Cell Differentiation , Cell Movement , Embryonic Development , MAP Kinase Signaling System , Animals , Female , Pregnancy , Blastocyst/metabolism , Blastocyst/cytology , Blastocyst Inner Cell Mass/metabolism , Blastocyst Inner Cell Mass/cytology , Cell Lineage , Sheep , Signal Transduction , Mice
6.
Mol Reprod Dev ; 91(5): e23760, 2024 May.
Article in English | MEDLINE | ID: mdl-38769918

ABSTRACT

e-Lysine acetylation is a prominent histone mark found at transcriptionally active loci. Among many lysine acetyl transferases, nonspecific lethal complex (NSL) members are known to mediate the modification of histone H4. In addition to histone modifications, the KAT8 regulatory complex subunit 3 gene (Kansl3), a core member of NSL complex, has been shown to be involved in several other cellular processes such as mitosis and mitochondrial activity. Although functional studies have been performed on NSL complex members, none of the four core proteins, including Kansl3, have been studied during early mouse development. Here we show that homozygous knockout Kansl3 embryos are lethal at peri-implantation stages, failing to hatch out of the zona pellucida. When the zona pellucida is removed in vitro, Kansl3 null embryos form an abnormal outgrowth with significantly disrupted inner cell mass (ICM) morphology. We document lineage-specific defects at the blastocyst stage with significantly reduced ICM cell number but no difference in trophectoderm cell numbers. Both epiblast and primitive endoderm lineages are altered with reduced cell numbers in null mutants. These results show that Kansl3 is indispensable during early mouse embryonic development and with defects in both ICM and trophectoderm lineages.


Subject(s)
Embryonic Development , Animals , Female , Mice , Blastocyst/metabolism , Blastocyst/cytology , Blastocyst Inner Cell Mass/metabolism , Blastocyst Inner Cell Mass/cytology , Embryo Loss/pathology , Embryo Loss/genetics , Embryo Loss/metabolism , Embryonic Development/genetics , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/deficiency , Mice, Knockout
7.
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
8.
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
9.
J Reprod Dev ; 67(3): 161-165, 2021 Jun 21.
Article in English | MEDLINE | ID: mdl-33907058

ABSTRACT

In mammalian embryos, the first visible differentiation event is the segregation of the inner cell mass (ICM) and trophectoderm (TE) during the transition from the morula to the blastocyst stage. The ICM, which is attached to the inside of the TE, develop into the fetus and extraembryonic tissues, while the TE, which is a single layer surrounding the fluid-filled cavity called the blastocoel, will provide extraembryonic structures such as the placenta. ICM/TE differentiation is regulated by the interaction between various transcriptional factors. However, little information is available on the segregation of the ICM and TE lineages in preimplantation embryos of domestic animals, such as cattle and pigs. This review focuses on the roles of cell differentiation factors that regulate the ICM/TE segregation of preimplantation bovine and porcine embryos. Understanding the mechanism of cell differentiation in early embryos is necessary to improve the in vitro production systems for bovine and porcine embryos.


Subject(s)
Blastocyst/metabolism , Cell Differentiation/physiology , Embryonic Development/physiology , Transcription Factors/metabolism , Animals , Animals, Domestic , Blastocyst/cytology , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cattle , Female , Swine , Transcription Factors/genetics
10.
Proc Natl Acad Sci U S A ; 115(41): 10387-10391, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30257947

ABSTRACT

Following erasure in the blastocyst, the entire genome undergoes de novo methylation at the time of implantation, with CpG islands being protected from this process. This bimodal pattern is then preserved throughout development and the lifetime of the organism. Using mouse embryonic stem cells as a model system, we demonstrate that the binding of an RNA polymerase complex on DNA before de novo methylation is predictive of it being protected from this modification, and tethering experiments demonstrate that the presence of this complex is, in fact, sufficient to prevent methylation at these sites. This protection is most likely mediated by the recruitment of enzyme complexes that methylate histone H3K4 over a local region and, in this way, prevent access to the de novo methylation complex. The topological pattern of H3K4me3 that is formed while the DNA is as yet unmethylated provides a strikingly accurate template for modeling the genome-wide basal methylation pattern of the organism. These results have far-reaching consequences for understanding the relationship between RNA transcription and DNA methylation.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , DNA Methylation , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Transcription, Genetic , Animals , Blastocyst Inner Cell Mass/cytology , CpG Islands , DNA-Directed RNA Polymerases/metabolism , Embryo, Mammalian/cytology , Mice , Mice, Transgenic , Transcription Factors/metabolism
11.
Genes Dev ; 27(12): 1378-90, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23788624

ABSTRACT

Although it is known that OCT4-NANOG are required for maintenance of pluripotent cells in vitro, the upstream signals that regulate this circuit during early development in vivo have not been identified. Here we demonstrate, for the first time, signal transducers and activators of transcription 3 (STAT3)-dependent regulation of the OCT4-NANOG circuitry necessary to maintain the pluripotent inner cell mass (ICM), the source of in vitro-derived embryonic stem cells (ESCs). We show that STAT3 is highly expressed in mouse oocytes and becomes phosphorylated and translocates to the nucleus in the four-cell and later stage embryos. Using leukemia inhibitory factor (Lif)-null embryos, we found that STAT3 phosphorylation is dependent on LIF in four-cell stage embryos. In blastocysts, interleukin 6 (IL-6) acts in an autocrine fashion to ensure STAT3 phosphorylation, mediated by janus kinase 1 (JAK1), a LIF- and IL-6-dependent kinase. Using genetically engineered mouse strains to eliminate Stat3 in oocytes and embryos, we firmly establish that STAT3 is essential for maintenance of ICM lineages but not for ICM and trophectoderm formation. Indeed, STAT3 directly binds to the Oct4 and Nanog distal enhancers, modulating their expression to maintain pluripotency of mouse embryonic and induced pluripotent stem cells. These results provide a novel genetic model of cell fate determination operating through STAT3 in the preimplantation embryo and pluripotent stem cells in vivo.


Subject(s)
Blastocyst Inner Cell Mass , Cell Lineage , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins , Octamer Transcription Factor-3 , STAT3 Transcription Factor , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphorylation , Pluripotent Stem Cells/physiology , Protein Binding , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
12.
Development ; 144(3): 519-528, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27993980

ABSTRACT

During development from oocyte to embryo, genetic programs in mouse germ cells are reshaped by chromatin remodeling to orchestrate the onset of development. Epigenetic modifications of specific amino acid residues of core histones and their isoforms can dramatically alter activation and suppression of gene expression. H3.3 is a histone H3 variant that plays essential roles in mouse oocytes and early embryos, but the functional role of individual amino acid residues has been unclear because of technical hurdles. Here, we describe two strategies that successfully investigated the functions of three individual H3.3 residues in oogenesis, cleavage-stage embryogenesis and early development. We first generated genetic mosaic ovaries and blastocysts with stochastic expression of wild-type or mutant H3.3 alleles and showed dominant negative effects of H3.3R26 and H3.3K27 in modulating oogenesis and partitioning cells to the inner cell mass of the early embryo. Time-lapse imaging assays also revealed the essential roles of H3.3K56 in efficient H2B incorporation and paternal pronuclei formation. Application of these strategies can be extended to investigate roles of additional H3.3 residues and has implications for use in other developmental systems.


Subject(s)
Blastocyst/metabolism , Histones/metabolism , Oocytes/metabolism , Animals , Blastocyst/cytology , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Blastomeres/cytology , Blastomeres/metabolism , Cell Lineage , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Female , Histones/chemistry , Histones/genetics , Male , Mice , Mice, Transgenic , Mosaicism , Oogenesis , Time-Lapse Imaging , Zygote/cytology , Zygote/metabolism
13.
Mol Hum Reprod ; 26(11): 866-878, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33010164

ABSTRACT

The preimplantation stage of development is exquisitely sensitive to environmental stresses, and changes occurring during this developmental phase may have long-term health effects. Animal studies indicate that IVF offspring display metabolic alterations, including hypertension, glucose intolerance and cardiac hypertrophy, often in a sexual dimorphic fashion. The detailed nature of epigenetic changes following in-vitro culture is, however, unknown. This study was performed to evaluate the epigenetic (using whole-genome bisulfite sequencing (WGBS) and assay for transposase-accessible chromatin using sequencing (ATAC-seq)) and transcriptomic changes (using RNA-seq) occurring in the inner cell mass (ICM) of male or female mouse embryos generated in vivo or by IVF. We found that the ICM of IVF embryos, compared to the in-vivo ICM, differed in 3% of differentially methylated regions (DMRs), of which 0.1% were located on CpG islands. ATAC-seq revealed that 293 regions were more accessible and 101 were less accessible in IVF embryos, while RNA-seq revealed that 21 genes were differentially regulated in IVF embryos. Functional enrichment analysis revealed that stress signalling (STAT and NF-kB signalling), developmental processes and cardiac hypertrophy signalling showed consistent changes in WGBS and ATAC-seq platforms. In contrast, male and female embryos showed minimal changes. Male ICM had an increased number of significantly hyper-methylated DMRs, while only 27 regions showed different chromatin accessibility and only one gene was differentially expressed. In summary, this study provides the first comprehensive analysis of DNA methylation, chromatin accessibility and RNA expression changes induced by IVF in male and female ICMs. This dataset can be of value to all researchers interested in the developmental origin of health and disease (DOHaD) hypothesis and might lead to a better understanding of how early embryonic manipulation may affect adult health.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , Epigenesis, Genetic/physiology , Sex Characteristics , Animals , Cells, Cultured , Chromatin/metabolism , CpG Islands , DNA Methylation , Embryo Culture Techniques , Embryo, Mammalian , Female , Fertilization/physiology , Fertilization in Vitro/methods , Fertilization in Vitro/veterinary , Gene Expression Profiling , Male , Mice , Pregnancy , Transcriptome
14.
Mol Hum Reprod ; 26(9): 702-711, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32663300

ABSTRACT

Early embryonic development is characterized by drastic changes in chromatin structure that affects the accessibility of the chromatin. In human, the chromosome reorganization and its involvement in the first linage segregation are poorly characterized due to the difficulties in obtaining human embryonic material and limitation on low input technologies. In this study, we aimed to explore the chromatin remodeling pattern in human preimplantation embryos and gain insight into the epigenetic regulation of inner cell mass (ICM) and trophectoderm (TE) differentiation. We optimized ATAC-seq (an assay for transposase-accessible chromatin using sequencing) to analyze the chromatin accessibility landscape for low DNA input. Sixteen preimplantation human blastocysts frozen on Day 6 were used. Our data showed that ATAC peak distributions of the promoter regions (<1 kb) and distal regions versus other regions were significantly different between ICM versus TE samples (P < 0.01). We detected that a higher percentage of accessible binding loci were located within 1 kb of the transcription start site in ICM compared to TE (P < 0.01). However, a higher percentage of accessible regions was detected in the distal region of TE compared to ICM (P < 0.01). In addition, eight differential peaks with a false discovery rate <0.05 between ICM and TE were detected. This is the first study to compare the landscape of the accessible chromatin between ICM and TE of human preimplantation embryos, which unveiled chromatin-level epigenetic regulation of cell lineage specification in early embryo development.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , Chromatin/metabolism , Ectoderm/metabolism , Adult , Blastocyst/chemistry , Blastocyst/metabolism , Blastocyst Inner Cell Mass/chemistry , Cells, Cultured , Chromatin/chemistry , Chromatin Assembly and Disassembly/physiology , DNA, Intergenic/analysis , DNA, Intergenic/metabolism , Ectoderm/chemistry , Embryonic Development/genetics , Female , Humans , Promoter Regions, Genetic/genetics
15.
Reproduction ; 159(1): 1-13, 2020 01.
Article in English | MEDLINE | ID: mdl-31671403

ABSTRACT

Microspherule protein 1 (MCRS1, also known as MSP58) is an evolutionarily conserved protein that has been implicated in various biological processes. Although a variety of functions have been attributed to MCRS1 in vitro, mammalian MCRS1 has not been studied in vivo. Here we report that MCRS1 is essential during early murine development. Mcrs1 mutant embryos exhibit normal morphology at the blastocyst stage but cannot be recovered at gastrulation, suggesting an implantation failure. Outgrowth (OG) assays reveal that mutant blastocysts do not form a typical inner cell mass (ICM) colony, the source of embryonic stem cells (ESCs). Surprisingly, cell death and histone H4 acetylation analysis reveal that apoptosis and global H4 acetylation are normal in mutant blastocysts. However, analysis of lineage specification reveals that while the trophoblast and primitive endoderm are properly specified, the epiblast lineage is compromised and exhibits a severe reduction in cell number. In summary, our study demonstrates the indispensable role of MCRS1 in epiblast development during early mammalian embryogenesis.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Embryo, Mammalian/cytology , Embryonic Development , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mutation , RNA-Binding Proteins/physiology , Animals , Blastocyst Inner Cell Mass/metabolism , Cell Differentiation , Cell Lineage , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Germ Layers/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Stem Cells ; 37(7): 888-898, 2019 07.
Article in English | MEDLINE | ID: mdl-30913328

ABSTRACT

Receptor tyrosine kinase signaling pathways are key regulators for the formation of the primitive endoderm (PrE) and the epiblast (Epi) from the inner cell mass (ICM) of the mouse preimplantation embryo. Among them, FGF signaling is critical for PrE cell specification, whereas PDGF signaling is critical for the survival of committed PrE cells. Here, we investigated possible functional redundancies among FGF, PDGF, and KIT signaling and showed that only PDGF signaling is involved in PrE cell survival. In addition, we analyzed the effectors downstream of PDGFRα. Our results suggest that the role of PDGF signaling in PrE cell survival is mediated through PI3K-mTOR and independently from p53. Lastly, we uncovered a role for PI3K-mTOR signaling in the survival of Epi cells. Taken together, we propose that survival of ICM cell lineages relies on the regulation of PI3K-mTOR signaling through the regulation of multiple signaling pathways. Stem Cells 2019;37:888-898.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , Cell Lineage/genetics , Endoderm/metabolism , Gene Expression Regulation, Developmental , Phosphatidylinositol 3-Kinases/genetics , Platelet-Derived Growth Factor/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Blastocyst , Blastocyst Inner Cell Mass/cytology , Cell Survival , Endoderm/cytology , Endoderm/growth & development , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
J Assist Reprod Genet ; 37(3): 559-571, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32112203

ABSTRACT

PURPOSE: To assess the accuracy and reliability of comprehensive chromosome screening by next-generation sequencing (NGS) of human trophectoderm (TE) biopsy specimens. METHODS: The reliability and accuracy of diagnoses made by preimplantation genetic testing for aneuploidy (PGT-A) from TE biopsy were tested. Repeat biopsies of TE and inner cell mass (ICM) samples were obtained from thawed blastocysts previously tested by NGS. To test for the reliability of the NGS assay, biopsy samples were compared with the original PGT-A results. Prior NGS testing classified the TE samples as euploid, aneuploid, or aneuploid-mosaic. The resulting re-biopsied samples underwent SurePlex whole genome amplification followed by NGS via the MiSeq platform, with copy number value (CNV) determined using BlueFuse Multi Software. The primary outcome measure was reliability, defined as concordance between initial TE result and the repeat biopsies. Accuracy was determined by concordance between the TE and ICM samples, and compared between three chromosome types (disomic, aneuploid, and mosaic). RESULTS: Re-biopsies were performed on 32 embryos with prior PGT-A showing euploidy (10 embryos), aneuploidy of one or two chromosomes (4 embryos), or aneuploid-mosaic with one aneuploid chromosome and one mosaic chromosome (18 embryos). One hundred twenty-nine biopsy samples completed NGS (90 TE and 39 ICM biopsies) and 105 biopsy results were included in the analysis. TE biopsies provide a highly accurate test of the future fetus, with the ICM disomic concordance rate of 97.6%. Clinical concordance rates indicate that TE biopsies provide a reliable test when the result is euploid (99.5%) or aneuploid (97.3%), but less reliable when the result is mosaic (35.2%). CONCLUSION: TE biopsies predict euploidy or aneuploidy in the ICM with a high degree of accuracy. PGT-A with NGS of TE biopsies is shown to be highly reliable, with clinically relevant concordance rates for aneuploidy and euploidy over 95%. TE biopsies indicating mosaicism were less reliable (35.2%), presumably because mitotic non-disjunction events are not uniformly distributed throughout the blastocyst. However, classification of TE biopsy of PGT-A with NGS results as either aneuploid or euploid provides a highly reliable test.


Subject(s)
Chromosomes/genetics , Genetic Testing , Mosaicism , Preimplantation Diagnosis , Adult , Aneuploidy , Biopsy , Blastocyst/metabolism , Blastocyst Inner Cell Mass/metabolism , Blastocyst Inner Cell Mass/pathology , Ectoderm/growth & development , Ectoderm/metabolism , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Pilot Projects , Pregnancy
18.
Dev Biol ; 442(1): 155-161, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30026121

ABSTRACT

FGF signaling is known to play a critical role in the specification of primitive endoderm (PrE) and epiblast (Epi) from the inner cell mass (ICM) during mouse preimplantation development, but how FGFs synergize with other growth factor signaling pathways is unknown. Because PDGFRα signaling has also been implicated in the PrE, we investigated the coordinate functions of PDGFRα together with FGFR1 or FGFR2 in PrE development. PrE development was abrogated in Pdgfra; Fgfr1 compound mutants, or significantly reduced in Pdgfra; Fgfr2 or PdgfraPI3K; Fgfr2 compound mutants. We provide evidence that both Fgfr2 and Pdgfra play roles in PrE cell survival while Fgfr1 controls PrE cell specification. Our results suggest a model where FGFR1-engaged ERK1/2 signaling governs PrE specification while PDGFRα- and by analogy possibly FGFR2- engaged PI3K signaling regulates PrE survival and positioning in the embryo. Together, these studies indicate how multiple growth factors and signaling pathways can cooperate in preimplantation development.


Subject(s)
Fibroblast Growth Factor 4/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Blastocyst/metabolism , Blastocyst Inner Cell Mass/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Embryo, Mammalian/metabolism , Embryonic Development , Endoderm/metabolism , Fibroblast Growth Factor 4/physiology , Fibroblast Growth Factors/metabolism , Germ Layers/metabolism , MAP Kinase Signaling System/physiology , Mice , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/physiology
19.
Hum Reprod ; 34(9): 1746-1761, 2019 09 29.
Article in English | MEDLINE | ID: mdl-31419301

ABSTRACT

STUDY QUESTION: Can miRNAs be reliably detected in the spent blastocyst media (SBM) after IVF as putative biomarkers of the implantation potential of euploid embryos? SUMMARY ANSWER: Adjustment of the data for blastocyst quality and the day of full-expansion hinders the predictive power of a fast, inexpensive, reproducible and user-friendly protocol based on the detection of 10 selected miRNAs from SBM. WHAT IS KNOWN ALREADY: Euploidy represents so far the strongest predictor of blastocyst competence. Nevertheless, ~50% of the euploid blastocysts fail to implant. Several studies across the years have suggested that a dialogue exists between the embryo and the endometrium aimed at the establishment of a pregnancy. MicroRNAs have been proposed as mediators of such a dialogue and investigated in this respect. Several expensive, time-consuming and complex protocols have been adopted and promising results have been produced, but conclusive evidence from large clinical studies is missing. STUDY DESIGN, SIZE, DURATION: This study was conducted in two phases from September 2015 to December 2017. In Phase 1, the human blastocyst miRNome profile was defined from the inner cell mass (ICM) and the corresponding whole-trophectoderm (TE) of six donated blastocysts. Two different protocols were adopted to this end. In parallel, 6 pools of 10 SBM each were run (3 from only implanted euploid blastocysts, IEBs; and 3 from only not-implanted euploid blastocysts, not-IEBs). A fast, inexpensive and user-friendly custom protocol for miRNA SBM profiling was designed. In Phase 2, 239 SBM from IEB and not-IEB were collected at three IVF centres. After 18 SBM from poor-quality blastocysts were excluded from the analysis, data from 107 SBM from not-IEB and 114 from IEB were produced through the previously developed custom protocol and compared. The data were corrected through logistic regressions. PARTICIPANT/MATERIALS, SETTINGS, METHODS: Donated blastocysts underwent ICM and whole-TE isolation. SBM were collected during IVF cycles characterized by ICSI, blastocyst culture in a continuous media, TE biopsy without zona pellucida opening in Day 3, quantitative PCR (qPCR)-based aneuploidy testing and vitrified-warmed single euploid embryo transfer. Not-IEB and IEB were clustered following a negative pregnancy test and a live birth, respectively. The Taqman Low Density Array (TLDA) cards and the Exiqon microRNA human panel I+II qPCR analysis protocols were adopted to analyse the ICM and whole-TE. The latter was used also for SBM pools. A custom protocol and plate was then designed based on the Exiqon workflow, validated and finally adopted for SBM analysis in study Phase 2. This custom protocol allows the analysis of 10 miRNAs from 10 SBM in 3 hours from sample collection to data inspection. MAIN RESULTS AND ROLE OF THE CHANCE: The TLDA cards protocol involved a higher rate of false positive results (5.6% versus 2.8% with Exiqon). There were 44 miRNAs detected in the ICM and TE from both the protocols. One and 24 miRNAs were instead detected solely in the ICM and the TE, respectively. Overall, 29 miRNAs were detected in the pooled SBM: 8 only from not-IEB, 8 only from IEB and 13 from both. Most of them (N = 24/29, 82.7%) were also detected previously in both the ICM and TE with the Exiqon protocol; two miRNAs (N = 2/29, 6.9%) were previously detected only in the TE, and three (N = 3/29, 10.3%) were never detected previously. In study Phase 2, significant differences were shown between not-IEB and IEB in terms of both miRNA detection and relative quantitation. However, when the data were corrected for embryo morphology and day of full development (i.e. SBM collection), no significant association was confirmed. LIMITATIONS, REASONS FOR CAUTION: This study did not evaluate specifically exosomal miRNAs, thereby reducing the chance of identifying the functional miRNAs. Ex-vivo experiments are required to confirm the role of miRNAs in mediating the dialogue with endometrial cells, and higher throughput technologies need to be further evaluated for miRNA profiling from clinical SBM samples. WIDER IMPLICATIONS OF THE FINDINGS: Although no clinical predictive power was reported in this study, the absence of invasiveness related with SBM analysis and the evidence that embryonic genetic material can be reliably detected and analysed from SBM make this waste product of IVF an important source for further investigations aimed at improving embryo selection. STUDY FUNDING/COMPETING INTEREST(S): This project has been financially supported by Merck KgaA (Darmstadt, Germany) with a Grant for Fertility Innovation (GFI) 2015. The authors have no conflict of interest to declare related with this study. TRIAL REGISTRATION NUMBER: None.


Subject(s)
Aneuploidy , Blastocyst Inner Cell Mass/metabolism , Culture Media/chemistry , Embryo Culture Techniques/methods , Embryo Implantation , Fertilization in Vitro/methods , MicroRNAs/genetics , Preimplantation Diagnosis/methods , Adult , Biomarkers , Female , Humans , Middle Aged , Polymerase Chain Reaction , Pregnancy , Preimplantation Diagnosis/economics , Reproducibility of Results , Single Embryo Transfer , Vitrification
20.
Reproduction ; 158(4): 303-312, 2019 10.
Article in English | MEDLINE | ID: mdl-31408846

ABSTRACT

Supplementing interleukin-6 (IL6) to in vitro-produced bovine embryos increases inner cell mass (ICM) cell numbers in blastocysts. A series of studies were completed to further dissect this effect. Treatment with IL6 increased ICM cell numbers in early, regular and expanded blastocysts but had no effect on morulae total cell number. Treatment with IL6 for 30 min induced signal transducer and activator of transcription 3 (STAT3) phosphorylation and nuclear translocation in all blastomeres in early morulae and specifically within the ICM in blastocysts. Also, IL6 supplementation increased SOCS3 mRNA abundance, a STAT3-responsive gene, in blastocysts. Chemical inhibition of Janus kinase (JAK) activity from day 5 to day 8 prevented STAT3 activation and the IL6-induced ICM cell number increase. Global transcriptome analysis of blastocysts found that transcripts for IL6 and its receptor subunits (IL6R and IL6ST) were the most abundantly expressed IL6 family ligand and receptors. These results indicate that IL6 increases ICM cell numbers as the ICM lineage emerges at the early blastocyst stage through a STAT3-dependent mechanism. Also, IL6 appears to be the primary IL6 cytokine family member utilized by bovine blastocysts to control ICM cell numbers.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Blastomeres/cytology , Interleukin-6/metabolism , Janus Kinases/metabolism , Morula/cytology , STAT3 Transcription Factor/metabolism , Animals , Blastocyst Inner Cell Mass/metabolism , Blastomeres/metabolism , Cattle , Female , Morula/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL