Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 317
Filter
1.
Cell ; 183(6): 1665-1681.e18, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33188776

ABSTRACT

We present deterministic barcoding in tissue for spatial omics sequencing (DBiT-seq) for co-mapping of mRNAs and proteins in a formaldehyde-fixed tissue slide via next-generation sequencing (NGS). Parallel microfluidic channels were used to deliver DNA barcodes to the surface of a tissue slide, and crossflow of two sets of barcodes, A1-50 and B1-50, followed by ligation in situ, yielded a 2D mosaic of tissue pixels, each containing a unique full barcode AB. Application to mouse embryos revealed major tissue types in early organogenesis as well as fine features like microvasculature in a brain and pigmented epithelium in an eye field. Gene expression profiles in 10-µm pixels conformed into the clusters of single-cell transcriptomes, allowing for rapid identification of cell types and spatial distributions. DBiT-seq can be adopted by researchers with no experience in microfluidics and may find applications in a range of fields including developmental biology, cancer biology, neuroscience, and clinical pathology.


Subject(s)
DNA Barcoding, Taxonomic , Genomics , Organ Specificity/genetics , Animals , Automation , Brain/embryology , Cluster Analysis , DNA, Complementary/genetics , Embryo, Mammalian/metabolism , Eye/embryology , Female , Gene Expression Regulation, Developmental , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Microfluidics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Single-Cell Analysis , Transcriptome/genetics
2.
Cell ; 173(3): 776-791.e17, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29576449

ABSTRACT

Transformation from morula to blastocyst is a defining event of preimplantation embryo development. During this transition, the embryo must establish a paracellular permeability barrier to enable expansion of the blastocyst cavity. Here, using live imaging of mouse embryos, we reveal an actin-zippering mechanism driving this embryo sealing. Preceding blastocyst stage, a cortical F-actin ring assembles at the apical pole of the embryo's outer cells. The ring structure forms when cortical actin flows encounter a network of polar microtubules that exclude F-actin. Unlike stereotypical actin rings, the actin rings of the mouse embryo are not contractile, but instead, they expand to the cell-cell junctions. Here, they couple to the junctions by recruiting and stabilizing adherens and tight junction components. Coupling of the actin rings triggers localized myosin II accumulation, and it initiates a tension-dependent zippering mechanism along the junctions that is required to seal the embryo for blastocyst formation.


Subject(s)
Actins/chemistry , Blastocyst/metabolism , Microtubules/metabolism , Myosin Type II/chemistry , Animals , Cell Communication , Cytoskeletal Proteins/chemistry , Embryo, Mammalian , Embryonic Development , Female , Green Fluorescent Proteins , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Morula , RNA, Small Interfering/metabolism , Tight Junctions
3.
Genes Dev ; 37(9-10): 418-431, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37257918

ABSTRACT

Translation of maternal mRNAs is detected before transcription of zygotic genes and is essential for mammalian embryo development. How certain maternal mRNAs are selected for translation instead of degradation and how this burst of translation affects zygotic genome activation remain unknown. Using gene-edited mice, we document that the oocyte-specific eukaryotic translation initiation factor 4E family member 1b (eIF4E1b) is the regulator of maternal mRNA expression that ensures subsequent reprogramming of the zygotic genome. In oocytes, eIF4E1b binds to transcripts encoding translation machinery proteins, chromatin remodelers, and reprogramming factors to promote their translation in zygotes and protect them from degradation. The protein products are thought to establish an open chromatin landscape in one-cell zygotes to enable transcription of genes required for cleavage stage development. Our results define a program for rapid resetting of the zygotic epigenome that is regulated by maternal mRNA expression and provide new insights into the mammalian maternal-to-zygotic transition.


Subject(s)
RNA, Messenger, Stored , Zygote , Animals , Mice , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Oocytes , Protein Biosynthesis , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/metabolism , Zygote/metabolism
4.
Annu Rev Cell Dev Biol ; 32: 677-691, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27298093

ABSTRACT

The two vascular systems of our body are the blood and the lymphatic vasculature. Our understanding of the genes and molecular mechanisms controlling the development of the lymphatic vasculature network has significantly improved. The availability of novel animal models and better imaging tools led to the identification of lymphatics in tissues and organs previously thought to be devoid of them. Similarly, the classical textbook list of established functional roles of the lymphatic system has been expanded by the addition of novel findings. In this review we provide a historical perspective of some of the important landmarks that opened the doors to researchers working in this field. We also summarize some of the current views about embryonic lymphangiogenesis, particularly about the source(s), commitment, and differentiation of lymphatic endothelial cells.


Subject(s)
Cell Lineage , Lymphangiogenesis , Animals , Blood Vessels/physiology , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans
5.
Genes Dev ; 36(7-8): 483-494, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35483741

ABSTRACT

Genomic imprinting regulates parental origin-dependent monoallelic gene expression. It is mediated by either germline differential methylation of DNA (canonical imprinting) or oocyte-derived H3K27me3 (noncanonical imprinting) in mice. Depletion of Eed, an essential component of Polycomb repressive complex 2, results in genome-wide loss of H3K27me3 in oocytes, which causes loss of noncanonical imprinting (LOI) in embryos. Although Eed maternal KO (matKO) embryos show partial lethality after implantation, it is unknown whether LOI itself contributes to the developmental phenotypes of these embryos, which makes it unclear whether noncanonical imprinting is developmentally relevant. Here, by combinatorial matKO of Xist, a noncanonical imprinted gene whose LOI causes aberrant transient maternal X-chromosome inactivation (XCI) at preimplantation, we show that prevention of the transient maternal XCI greatly restores the development of Eed matKO embryos. Moreover, we found that the placentae of Eed matKO embryos are remarkably enlarged in a manner independent of Xist LOI. Heterozygous deletion screening of individual autosomal noncanonical imprinted genes suggests that LOI of the Sfmbt2 miRNA cluster chromosome 2 miRNA cluster (C2MC), solute carrier family 38 member 4 (Slc38a4), and Gm32885 contributes to the placental enlargement. Taken together, our study provides evidence that Xist imprinting sustains embryonic development and that autosomal noncanonical imprinting restrains placental overgrowth.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Embryonic Development/genetics , Female , Histones/metabolism , Mice , Placenta , Pregnancy , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , X Chromosome Inactivation
6.
Genes Dev ; 2022 Aug 18.
Article in English | MEDLINE | ID: mdl-35981753

ABSTRACT

Promoter-proximal RNA Pol II pausing is a critical step in transcriptional control. Pol II pausing has been predominantly studied in tissue culture systems. While Pol II pausing has been shown to be required for mammalian development, the phenotypic and mechanistic details of this requirement are unknown. Here, we found that loss of Pol II pausing stalls pluripotent state transitions within the epiblast of the early mouse embryo. Using Nelfb -/- mice and a NELFB degron mouse pluripotent stem cell model, we show that embryonic stem cells (ESCs) representing the naïve state of pluripotency successfully initiate a transition program but fail to balance levels of induced and repressed genes and enhancers in the absence of NELF. We found an increase in chromatin-associated NELF during transition from the naïve to later pluripotent states. Overall, our work defines the acute and long-term molecular consequences of NELF loss and reveals a role for Pol II pausing in the pluripotency continuum as a modulator of cell state transitions.

7.
Annu Rev Genet ; 54: 167-187, 2020 11 23.
Article in English | MEDLINE | ID: mdl-32867543

ABSTRACT

Cellular heterogeneity is a property of any living system; however, its relationship with cellular fate decision remains an open question. Recent technological advances have enabled valuable insights, especially in complex systems such as the mouse embryo. In this review, we discuss recent studies that characterize cellular heterogeneity at different levels during mouse development, from the two-cell stage up to gastrulation. In addition to key experimental findings, we review mathematical modeling approaches that help researchers interpret these findings. Disentangling the role of heterogeneity in cell fate decision will likely rely on the refined integration of experiments, large-scale omics data, and mathematical modeling, complemented by the use of synthetic embryos and gastruloids as promising in vitro models.


Subject(s)
Embryo, Mammalian/physiology , Animals , Cell Differentiation/physiology , Humans , Mice
8.
EMBO J ; 41(7): e108747, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35266581

ABSTRACT

Mesoderm arises at gastrulation and contributes to both the mouse embryo proper and its extra-embryonic membranes. Two-photon live imaging of embryos bearing a keratin reporter allowed recording filament nucleation and elongation in the extra-embryonic region. Upon separation of amniotic and exocoelomic cavities, keratin 8 formed apical cables co-aligned across multiple cells in the amnion, allantois, and blood islands. An influence of substrate rigidity and composition on cell behavior and keratin content was observed in mesoderm explants. Embryos lacking all keratin filaments displayed a deflated extra-embryonic cavity, a narrow thick amnion, and a short allantois. Single-cell RNA sequencing of sorted mesoderm cells and micro-dissected amnion, chorion, and allantois, provided an atlas of transcriptomes with germ layer and regional information. It defined the cytoskeleton and adhesion expression profile of mesoderm-derived keratin 8-enriched cells lining the exocoelomic cavity. Those findings indicate a novel role for keratin filaments in the expansion of extra-embryonic structures and suggest mechanisms of mesoderm adaptation to the environment.


Subject(s)
Gastrulation , Mesoderm , Animals , Embryo, Mammalian , Extraembryonic Membranes , Keratins/genetics , Keratins/metabolism , Mesoderm/metabolism , Mice
9.
Development ; 150(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-36994838

ABSTRACT

Transcriptional networks governing cardiac precursor cell (CPC) specification are incompletely understood owing, in part, to limitations in distinguishing CPCs from non-cardiac mesoderm in early gastrulation. We leveraged detection of early cardiac lineage transgenes within a granular single-cell transcriptomic time course of mouse embryos to identify emerging CPCs and describe their transcriptional profiles. Mesp1, a transiently expressed mesodermal transcription factor, is canonically described as an early regulator of cardiac specification. However, we observed perdurance of CPC transgene-expressing cells in Mesp1 mutants, albeit mislocalized, prompting us to investigate the scope of the role of Mesp1 in CPC emergence and differentiation. Mesp1 mutant CPCs failed to robustly activate markers of cardiomyocyte maturity and crucial cardiac transcription factors, yet they exhibited transcriptional profiles resembling cardiac mesoderm progressing towards cardiomyocyte fates. Single-cell chromatin accessibility analysis defined a Mesp1-dependent developmental breakpoint in cardiac lineage progression at a shift from mesendoderm transcriptional networks to those necessary for cardiac patterning and morphogenesis. These results reveal Mesp1-independent aspects of early CPC specification and underscore a Mesp1-dependent regulatory landscape required for progression through cardiogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Epigenomics , Myocytes, Cardiac , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Myocytes, Cardiac/metabolism , Transcription Factors/metabolism
10.
Semin Cell Dev Biol ; 138: 54-67, 2023 03 30.
Article in English | MEDLINE | ID: mdl-35277330

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a well-defined cellular process that was discovered in chicken embryos and described as "epithelial to mesenchymal transformation" [1]. During EMT, epithelial cells lose their epithelial features and acquire mesenchymal character with migratory potential. EMT has subsequently been shown to be essential for both developmental and pathological processes including embryo morphogenesis, wound healing, tissue fibrosis and cancer [2]. During the past 5 years, interest and study of EMT especially in cancer biology have increased exponentially due to the implied role of EMT in multiple aspects of malignancy such as cell invasion, survival, stemness, metastasis, therapeutic resistance and tumor heterogeneity [3]. Since the process of EMT in embryogenesis and cancer progression shares similar phenotypic changes, core transcription factors and molecular mechanisms, it has been proposed that the initiation and development of carcinoma could be attributed to abnormal activation of EMT factors usually required for normal embryo development. Therefore, developmental EMT mechanisms, whose timing, location, and tissue origin are strictly regulated, could prove useful for uncovering new insights into the phenotypic changes and corresponding gene regulatory control of EMT under pathological conditions. In this review, we initially provide an overview of the phenotypic and molecular mechanisms involved in EMT and discuss the newly emerging concept of epithelial to mesenchymal plasticity (EMP). Then we focus on our current knowledge of a classic developmental EMT event, neural crest cell (NCC) delamination, highlighting key differences in our understanding of NCC EMT between mammalian and non-mammalian species. Lastly, we highlight available tools and future directions to advance our understanding of mammalian NCC EMT.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Animals , Chick Embryo , Epithelial-Mesenchymal Transition/genetics , Neural Crest , Cell Adhesion , Embryonic Development/genetics , Neoplasms/pathology , Mammals
11.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35037942

ABSTRACT

Generating comprehensive image maps, while preserving spatial three-dimensional (3D) context, is essential in order to locate and assess quantitatively specific cellular features and cell-cell interactions during organ development. Despite recent advances in 3D imaging approaches, our current knowledge of the spatial organization of distinct cell types in the embryonic pancreatic tissue is still largely based on two-dimensional histological sections. Here, we present a light-sheet fluorescence microscopy approach to image the pancreas in three dimensions and map tissue interactions at key time points in the mouse embryo. We demonstrate the utility of the approach by providing volumetric data, 3D distribution of three main cellular components (epithelial, mesenchymal and endothelial cells) within the developing pancreas, and quantification of their relative cellular abundance within the tissue. Interestingly, our 3D images show that endocrine cells are constantly and increasingly in contact with endothelial cells forming small vessels, whereas the interactions with mesenchymal cells decrease over time. These findings suggest distinct cell-cell interaction requirements for early endocrine cell specification and late differentiation. Lastly, we combine our image data in an open-source online repository (referred to as the Pancreas Embryonic Cell Atlas).


Subject(s)
Imaging, Three-Dimensional/methods , Pancreas/anatomy & histology , Animals , Embryo, Mammalian/anatomy & histology , Embryonic Development , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelium/anatomy & histology , Homeobox Protein Nkx-2.5/deficiency , Homeobox Protein Nkx-2.5/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence
12.
Proc Natl Acad Sci U S A ; 119(28): e2118182119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35787055

ABSTRACT

X chromosome inactivation (XCI) is the process of silencing one of the X chromosomes in cells of the female mammal which ensures dosage compensation between the sexes. Although theoretically random in somatic tissues, the choice of which X chromosome is chosen to be inactivated can be biased in mice by genetic element(s) associated with the so-called X-controlling element (Xce). Although the Xce was first described and genetically localized nearly 40 y ago, its mode of action remains elusive. In the approach presented here, we identify a single long noncoding RNA (lncRNA) within the Xce locus, Lppnx, which may be the driving factor in the choice of which X chromosome will be inactivated in the developing female mouse embryo. Comparing weak and strong Xce alleles we show that Lppnx modulates the expression of Xist lncRNA, one of the key factors in XCI, by controlling the occupancy of pluripotency factors at Intron1 of Xist. This effect is counteracted by enhanced binding of Rex1 in DxPas34, another key element in XCI regulating the activity of Tsix lncRNA, the main antagonist of Xist, in the strong but not in the weak Xce allele. These results suggest that the different susceptibility for XCI observed in weak and strong Xce alleles results from differential transcription factor binding of Xist Intron 1 and DxPas34, and that Lppnx represents a decisive factor in explaining the action of the Xce.


Subject(s)
RNA, Long Noncoding , X Chromosome Inactivation , Alleles , Animals , Dosage Compensation, Genetic , Female , Mammals/genetics , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome/genetics
13.
Genomics ; 116(3): 110836, 2024 05.
Article in English | MEDLINE | ID: mdl-38537809

ABSTRACT

The CRISPR/Cas9 system can induce off-target effects in programmed gene editing, but there have been few reports on cleavage detection and their affection in embryo development. To study these events, sgRNAs with different off-target rates were designed and compared after micro-injected into mouse zygotes, and γH2AX was used for DNA cleavage sites analysis by immunostaining and CUT&Tag. Although the low off-target sgRNA were usually selected for production gene editing animals, γH2AX immunofluorescence indicated that there was a relative DSB peak at 15 h after Cas9 system injection, and the number of γH2AX foci at the peak was significantly higher in the low off-target sgRNA-injected group than in the control group. Further, the result of CUT&Tag sequencing analysis showed more double-strand breaks (DSBs) related sequences were detected in low off-target sgRNA-injected group than control and the distribution of DSB related sequences had no chromosome specificity. Gene Ontology (GO) annotation analysis of the DSB related sequences showed that these sequences were mainly concentrated at genes associated with some important biological processes, molecular functions, and cell components. In a conclusion, there are many sgRNA-sequence-independent DSBs in early mouse embryos when the Cas9 system is used for gene editing and the DSB related sequence could be detected and characterized in the genome. These results and method should also be considered in using or optimizing the Cas9 system.


Subject(s)
CRISPR-Cas Systems , DNA Breaks, Double-Stranded , Embryo, Mammalian , Gene Editing , RNA, Guide, CRISPR-Cas Systems , Animals , Mice , Embryo, Mammalian/metabolism , Gene Editing/methods , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , DNA Cleavage , Zygote/metabolism , Histones/metabolism , Histones/genetics , Female
14.
BMC Bioinformatics ; 25(1): 294, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242990

ABSTRACT

Mouse (Mus musculus) models have been heavily utilized in developmental biology research to understand mammalian embryonic development, as mice share many genetic, physiological, and developmental characteristics with humans. New explorations into the integration of temporal (stage-specific) and transcriptional (tissue-specific) data have expanded our knowledge of mouse embryo tissue-specific gene functions. To better understand the substantial impact of synonymous mutational variations in the cell-state-specific transcriptome on a tissue's codon and codon pair usage landscape, we have established a novel resource-Mouse Embryo Codon and Codon Pair Usage Tables (Mouse Embryo CoCoPUTs). This webpage not only offers codon and codon pair usage, but also GC, dinucleotide, and junction dinucleotide usage, encompassing four strains, 15 murine embryonic tissue groups, 18 Theiler stages, and 26 embryonic days. Here, we leverage Mouse Embryo CoCoPUTs and employ the use of heatmaps to depict usage changes over time and a comparison to human usage for each strain and embryonic time point, highlighting unique differences and similarities. The usage similarities found between mouse and human central nervous system data highlight the translation for projects leveraging mouse models. Data for this analysis can be directly retrieved from Mouse Embryo CoCoPUTs. This cutting-edge resource plays a crucial role in deciphering the complex interplay between usage patterns and embryonic development, offering valuable insights into variation across diverse tissues, strains, and stages. Its applications extend across multiple domains, with notable advantages for biotherapeutic development, where optimizing codon usage can enhance protein expression; one can compare strains, tissues, and mouse embryonic stages in one query. Additionally, Mouse Embryo CoCoPUTs holds great potential in the field of tissue-specific genetic engineering, providing insights for tailoring gene expression to specific tissues for targeted interventions. Furthermore, this resource may enhance our understanding of the nuanced connections between usage biases and tissue-specific gene function, contributing to the development of more accurate predictive models for genetic disorders.


Subject(s)
Transcriptome , Animals , Mice , Transcriptome/genetics , Embryo, Mammalian/metabolism , Humans , Embryonic Development/genetics , Codon Usage/genetics
15.
Semin Cell Dev Biol ; 129: 63-74, 2022 09.
Article in English | MEDLINE | ID: mdl-35577698

ABSTRACT

Cellular protrusions generated by the actin cytoskeleton are central to the process of building the body of the embryo. Problems with cellular protrusions underlie human diseases and syndromes, including implantation defects and pregnancy loss, congenital birth defects, and cancer. Cells use protrusive activity together with actin-myosin contractility to create an ordered body shape of the embryo. Here, I review how actin-rich protrusions are used by two major morphological cell types, epithelial and mesenchymal cells, during collective cell migration to sculpt the mouse embryo body. Pre-gastrulation epithelial collective migration of the anterior visceral endoderm is essential for establishing the anterior-posterior body axis. Gastrulation mesenchymal collective migration of the mesoderm wings is crucial for body elongation, and somite and heart formation. Analysis of mouse mutants with disrupted cellular protrusions revealed the key role of protrusions in embryonic morphogenesis and embryo survival. Recent technical approaches have allowed examination of the mechanisms that control cell and tissue movements in vivo in the complex 3D microenvironment of living mouse embryos. Advancing our understanding of protrusion-driven morphogenesis should provide novel insights into human developmental disorders and cancer metastasis.


Subject(s)
Actins , Embryonic Development , Actins/metabolism , Animals , Cell Movement , Cell Surface Extensions/metabolism , Endoderm , Female , Gastrulation , Humans , Mesoderm , Mice , Pregnancy
16.
J Biol Chem ; 299(9): 105130, 2023 09.
Article in English | MEDLINE | ID: mdl-37543366

ABSTRACT

Long noncoding RNAs (lncRNAs) are increasingly being recognized as modulators in various biological processes. However, due to their low expression, their systematic characterization is difficult to determine. Here, we performed transcript annotation by a newly developed computational pipeline, termed RNA-seq and small RNA-seq combined strategy (RSCS), in a wide variety of cellular contexts. Thousands of high-confidence potential novel transcripts were identified by the RSCS, and the reliability of the transcriptome was verified by analysis of transcript structure, base composition, and sequence complexity. Evidenced by the length comparison, the frequency of the core promoter and the polyadenylation signal motifs, and the locations of transcription start and end sites, the transcripts appear to be full length. Furthermore, taking advantage of our strategy, we identified a large number of endogenous retrovirus-associated lncRNAs, and a novel endogenous retrovirus-lncRNA that was functionally involved in control of Yap1 expression and essential for early embryogenesis was identified. In summary, the RSCS can generate a more complete and precise transcriptome, and our findings greatly expanded the transcriptome annotation for the mammalian community.


Subject(s)
Molecular Sequence Annotation , RNA, Long Noncoding , RNA-Seq , Animals , Embryonic Development/genetics , Mammals/embryology , Mammals/genetics , Molecular Sequence Annotation/methods , Promoter Regions, Genetic/genetics , Reproducibility of Results , Retroviridae/genetics , RNA, Long Noncoding/genetics , RNA-Seq/methods , Transcription Initiation Site , Transcriptome/genetics , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/metabolism
17.
Development ; 148(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33199445

ABSTRACT

Anterior mesoderm (AM) and definitive endoderm (DE) progenitors represent the earliest embryonic cell types that are specified during germ layer formation at the primitive streak (PS) of the mouse embryo. Genetic experiments indicate that both lineages segregate from Eomes-expressing progenitors in response to different Nodal signaling levels. However, the precise spatiotemporal pattern of the emergence of these cell types and molecular details of lineage segregation remain unexplored. We combined genetic fate labeling and imaging approaches with single-cell RNA sequencing (scRNA-seq) to follow the transcriptional identities and define lineage trajectories of Eomes-dependent cell types. Accordingly, all cells moving through the PS during the first day of gastrulation express Eomes AM and DE specification occurs before cells leave the PS from Eomes-positive progenitors in a distinct spatiotemporal pattern. ScRNA-seq analysis further suggested the immediate and complete separation of AM and DE lineages from Eomes-expressing cells as last common bipotential progenitor.


Subject(s)
Cell Lineage , Endoderm/cytology , Endoderm/metabolism , Gastrulation , Mesoderm/cytology , Mesoderm/metabolism , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mice , Models, Biological , Primitive Streak/embryology , Primitive Streak/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Time Factors , Transcription, Genetic
18.
Reprod Biomed Online ; 49(4): 104120, 2024 10.
Article in English | MEDLINE | ID: mdl-39084124

ABSTRACT

RESEARCH QUESTION: To what extent does the type and concentration of protein and the type of culture medium affect the sensitivity of the mouse embryo assay (MEA) to detect Triton X-100 (TX-100) in culture media? DESIGN: The effect of the concentration of bovine serum albumin (BSA) and human serum albumin (HSA) was assessed by supplementing media with 0.5 or 5 mg/ml. Potassium-supplemented simplex optimized medium (KSOM) and human tubal fluid (HTF) were used as complex and simple formulation media, respectively. Variables were combined, forming study groups where embryos were cultured in test media spiked with a sublethal TX-100 concentration. The conditions of greatest sensitivity were determined by statistical comparison of blastocyst formation rates and total cell counts between groups. RESULTS: Although all of the study groups showed equal capacity for sustaining proper embryo development, the reported sensitivity of the MEA differed between groups when subjected to TX-100. HTF conferred significantly greater sensitivity than KSOM regardless of the type and concentration of protein used, and medium supplementation with 5 mg/ml BSA rather than 0.5 mg/ml BSA resulted in significantly higher sensitivity regardless of the type of medium used. This increase in concentration also resulted in higher sensitivity when supplementing HTF with HSA. The BSA groups provided more sensitivity than their HSA counterparts, except for the KSOM + 0.5 mg/ml BSA group. Cell count analysis did not provide further significant conclusions. CONCLUSIONS: For TX-100 detection within culture medium, the type and concentration of protein and the type of culture medium have a direct effect on MEA sensitivity. These results could help to standardize the MEA protocol, and increase its ability to detect sublethal concentrations of embryotoxic substances, especially TX-100, thus avoiding possible clinical harmful effects.


Subject(s)
Culture Media , Embryo Culture Techniques , Embryonic Development , Octoxynol , Serum Albumin, Bovine , Octoxynol/pharmacology , Animals , Mice , Serum Albumin, Bovine/pharmacology , Embryo Culture Techniques/methods , Female , Embryonic Development/drug effects , Embryo, Mammalian/drug effects , Humans , Serum Albumin, Human/analysis
19.
J Assist Reprod Genet ; 41(6): 1605-1617, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38557805

ABSTRACT

PURPOSE: Unpredictable genetic modifications and chromosomal aberrations following CRISPR/Cas9 administration hamper the efficacy of germline editing. Repair events triggered by double-strand DNA breaks (DSBs) besides non-homologous end joining and repair template-driven homology-directed repair have been insufficiently investigated in mouse. In this work, we are the first to investigate the precise repair mechanisms triggered by parental-specific DSB induction in mouse for paternal mutational correction in the context of an infertility-related mutation. METHODS: We aimed to correct a paternal 22-nucleotide deletion in Plcz1, associated with lack of fertilisation in vitro, by administrating CRISPR/Cas9 components during intracytoplasmic injection of Plcz1-null sperm in wild-type oocytes combined with assisted oocyte activation. Through targeted next-generation sequencing, 77 injected embryos and 26 blastomeres from seven injected embryos were investigated. In addition, low-pass whole genome sequencing was successfully performed on 17 injected embryo samples. RESULTS: Repair mechanisms induced by two different CRISPR/Cas9 guide RNA (gRNA) designs were investigated. In 13.73% (7/51; gRNA 1) and 19.05% (4/21; gRNA 2) of the targeted embryos, only the wild-type allele was observed, of which the majority (85.71%; 6/7) showed integrity of the targeted chromosome. Remarkably, for both designs, only in one of these embryos (1/7; gRNA 1 and 1/4; gRNA2) could repair template use be detected. This suggests that alternative repair events have occurred. Next, various genetic events within the same embryo were detected after single-cell analysis of four embryos. CONCLUSION: Our results suggest the occurrence of mosaicism and complex repair events after CRISPR/Cas9 DSB induction where chromosomal integrity is predominantly contained.


Subject(s)
CRISPR-Cas Systems , DNA Breaks, Double-Stranded , Gene Editing , Animals , CRISPR-Cas Systems/genetics , Mice , Female , Gene Editing/methods , Male , Oocytes/growth & development , Oocytes/metabolism , Infertility/genetics , Infertility/therapy , Mutation/genetics , DNA Repair/genetics , Embryo, Mammalian , Sperm Injections, Intracytoplasmic/methods , RNA, Guide, CRISPR-Cas Systems/genetics , DNA End-Joining Repair/genetics
20.
Genes Dev ; 30(6): 611-21, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26980186

ABSTRACT

In mammals, epigenetic reprogramming, the acquisition and loss of totipotency, and the first cell fate decision all occur within a 3-d window after fertilization from the one-cell zygote to the formation of the blastocyst. These processes are poorly understood in molecular detail, yet this is an essential prerequisite to uncover principles of stem cells, chromatin biology, and thus regenerative medicine. A unique feature of preimplantation development is the drastic genome-wide changes occurring to nuclear architecture. From studying somatic and in vitro cultured embryonic stem cells (ESCs) it is becoming increasingly established that the three-dimensional (3D) positions of genomic loci relative to each other and to specific compartments of the nucleus can act on the regulation of gene expression, potentially driving cell fate. However, the functionality, mechanisms, and molecular characteristics of the changes in nuclear organization during preimplantation development are only now beginning to be unraveled. Here, we discuss the peculiarities of nuclear compartments and chromatin organization during mammalian preimplantation development in the context of the transition from totipotency to pluripotency.


Subject(s)
Cell Nucleus/metabolism , Embryonic Development/physiology , Animals , Cell Lineage , Chromatin/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Humans
SELECTION OF CITATIONS
SEARCH DETAIL