Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.488
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000535

ABSTRACT

The receptive phase of the uterus is marked by structural and functional maturation of the endometrium. During this limited time span, the blastocyst competency is superimposed on the receptive endometrium. It is a well-known fact that lipid signalling in early-stage pregnancy has a crucial role in successful embryogenesis. In our study, CD-1 mouse uteri after normal and in vitro fertilization (IVF) were investigated at 6.5, 8.5, and 10.5 days of pregnancy. Matrix-assisted laser desorption/ionization time-of-flight imaging mass spectrometry and liquid chromatography coupled tandem mass spectrometry were used for identification of phosphatidylcholine (PC) lipid structures. In the embryonal tissues, PC 32:0 and PC 34:0 were increased, while in the antemesometrial (AM) decidua the two 20:4-containing PCs, PC 36:4 and PC 38:4 were increased. In transferred uterus samples, higher expressions of PC 34:0, PC 34:1, PC 34:2, PC 36:1, and PC 36:2 in mesometrial decidua were seen, whereas the two 20:4-containing PCs, PC 36:4 and PC 38:4 showed increased expression in the AM and lateral decidua. This paper shows a significant spatio-temporal change in lipid metabolism during IVF procedures for the first time.


Subject(s)
Fertilization in Vitro , Phosphatidylcholines , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Female , Animals , Mice , Phosphatidylcholines/metabolism , Phosphatidylcholines/analysis , Fertilization in Vitro/methods , Pregnancy , Embryo, Mammalian/metabolism , Embryonic Development , Uterus/metabolism , Blastocyst/metabolism
2.
Nat Cell Biol ; 26(7): 1200-1211, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38977846

ABSTRACT

Organogenesis is a highly complex and precisely regulated process. Here we profiled the chromatin accessibility in >350,000 cells derived from 13 mouse embryos at four developmental stages from embryonic day (E) 10.5 to E13.5 by SPATAC-seq in a single experiment. The resulting atlas revealed the status of 830,873 candidate cis-regulatory elements in 43 major cell types. By integrating the chromatin accessibility atlas with the previous transcriptomic dataset, we characterized cis-regulatory sequences and transcription factors associated with cell fate commitment, such as Nr5a2 in the development of gastrointestinal tract, which was preliminarily supported by the in vivo experiment in zebrafish. Finally, we integrated this atlas with the previous single-cell chromatin accessibility dataset from 13 adult mouse tissues to delineate the developmental stage-specific gene regulatory programmes within and across different cell types and identify potential molecular switches throughout lineage development. This comprehensive dataset provides a foundation for exploring transcriptional regulation in organogenesis.


Subject(s)
Chromatin , Gene Expression Regulation, Developmental , Organogenesis , Single-Cell Analysis , Zebrafish , Animals , Organogenesis/genetics , Chromatin/metabolism , Chromatin/genetics , Zebrafish/genetics , Zebrafish/embryology , Zebrafish/metabolism , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Lineage/genetics , Transcriptome/genetics , Embryo, Mammalian/metabolism , Female , Mice, Inbred C57BL
3.
Nat Commun ; 15(1): 6048, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025895

ABSTRACT

With the flourishing of spatial omics technologies, alignment and stitching of slices becomes indispensable to decipher a holistic view of 3D molecular profile. However, existing alignment and stitching methods are unpractical to process large-scale and image-based spatial omics dataset due to extreme time consumption and unsatisfactory accuracy. Here we propose SANTO, a coarse-to-fine method targeting alignment and stitching tasks for spatial omics. SANTO firstly rapidly supplies reasonable spatial positions of two slices and identifies the overlap region. Then, SANTO refines the positions of two slices by considering spatial and omics patterns. Comprehensive experiments demonstrate the superior performance of SANTO over existing methods. Specifically, SANTO stitches cross-platform slices for breast cancer samples, enabling integration of complementary features to synergistically explore tumor microenvironment. SANTO is then applied to 3D-to-3D spatiotemporal alignment to study development of mouse embryo. Furthermore, SANTO enables cross-modality alignment of spatial transcriptomic and epigenomic data to understand complementary interactions.


Subject(s)
Breast Neoplasms , Animals , Mice , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Transcriptome/genetics , Tumor Microenvironment/genetics , Epigenomics/methods , Genomics/methods , Algorithms , Embryo, Mammalian/metabolism , Imaging, Three-Dimensional/methods
4.
Commun Biol ; 7(1): 754, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38906986

ABSTRACT

Mammalian reproduction relies on precise maternal-fetal communication, wherein immune modifications foster tolerance toward the semi-allogeneic embryo. Extracellular vesicles (EVs), including exosomes and microvesicles, have emerged as crucial mediators, transporting molecules like microRNAs securely. EVs influence various reproductive stages, from gamete maturation to implantation, and impact pathologies like pregnancy loss. In the embryo-maternal dialogue, EVs notably affect oviductal interactions, gene expression, and the embryo-endometrial interface, crucial for successful implantation. Key queries persist about EV uptake, cargo delivery, and the specific biomolecules driving communication. Their potential in diagnostics, therapeutics, and understanding environmental impacts on fertility signals an exciting future, reliant on collaborative efforts for transformative strides in reproductive health.


Subject(s)
Extracellular Vesicles , Extracellular Vesicles/metabolism , Extracellular Vesicles/physiology , Humans , Female , Pregnancy , Animals , Maternal-Fetal Exchange , Embryo, Mammalian/metabolism , Cell Communication , Embryo Implantation , MicroRNAs/metabolism , MicroRNAs/genetics
5.
Cell Rep ; 43(6): 114372, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38878289

ABSTRACT

Emerging evidence highlights the regulatory role of paired-like (PRD-like) homeobox transcription factors (TFs) in embryonic genome activation (EGA). However, the majority of PRD-like genes are lost in rodents, thus prompting an investigation into PRD-like TFs in other mammals. Here, we showed that PRD-like TFs were transiently expressed during EGA in human, monkey, and porcine fertilized embryos, yet they exhibited inadequate expression in their cloned embryos. This study, using pig as the research model, identified LEUTX as a key PRD-like activator of porcine EGA through genomic profiling and found that LEUTX overexpression restored EGA failure and improved preimplantation development and cloning efficiency in porcine cloned embryos. Mechanistically, LEUTX opened EGA-related genomic regions and established histone acetylation via recruiting acetyltransferases p300 and KAT2A. These findings reveal the regulatory mechanism of LEUTX to govern EGA in pigs, which may provide valuable insights into the study of early embryo development for other non-rodent mammals.


Subject(s)
Genome , Nuclear Transfer Techniques , Animals , Swine , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Embryonic Development/genetics , Embryo, Mammalian/metabolism , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Acetylation , Cloning, Organism/methods , Histones/metabolism , Blastocyst/metabolism
6.
Nat Cell Biol ; 26(6): 962-974, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839978

ABSTRACT

Dynamic epigenomic reprogramming occurs during mammalian oocyte maturation and early development. However, the underlying transcription circuitry remains poorly characterized. By mapping cis-regulatory elements using H3K27ac, we identified putative enhancers in mouse oocytes and early embryos distinct from those in adult tissues, enabling global transitions of regulatory landscapes around fertilization and implantation. Gene deserts harbour prevalent putative enhancers in fully grown oocytes linked to oocyte-specific genes and repeat activation. Embryo-specific enhancers are primed before zygotic genome activation and are restricted by oocyte-inherited H3K27me3. Putative enhancers in oocytes often manifest H3K4me3, bidirectional transcription, Pol II binding and can drive transcription in STARR-seq and a reporter assay. Finally, motif analysis of these elements identified crucial regulators of oogenesis, TCF3 and TCF12, the deficiency of which impairs activation of key oocyte genes and folliculogenesis. These data reveal distinctive regulatory landscapes and their interacting transcription factors that underpin the development of mammalian oocytes and early embryos.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Oocytes , Oogenesis , Animals , Oocytes/metabolism , Female , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Oogenesis/genetics , Mice , Histones/metabolism , Histones/genetics , Embryo, Mammalian/metabolism , Mice, Inbred C57BL , Embryonic Development/genetics , Ovarian Follicle/metabolism , Mice, Knockout
7.
Nat Cell Biol ; 26(6): 868-877, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38849542

ABSTRACT

Despite a distinct developmental origin, extraembryonic cells in mice contribute to gut endoderm and converge to transcriptionally resemble their embryonic counterparts. Notably, all extraembryonic progenitors share a non-canonical epigenome, raising several pertinent questions, including whether this landscape is reset to match the embryonic regulation and if extraembryonic cells persist into later development. Here we developed a two-colour lineage-tracing strategy to track and isolate extraembryonic cells over time. We find that extraembryonic gut cells display substantial memory of their developmental origin including retention of the original DNA methylation landscape and resulting transcriptional signatures. Furthermore, we show that extraembryonic gut cells undergo programmed cell death and neighbouring embryonic cells clear their remnants via non-professional phagocytosis. By midgestation, we no longer detect extraembryonic cells in the wild-type gut, whereas they persist and differentiate further in p53-mutant embryos. Our study provides key insights into the molecular and developmental fate of extraembryonic cells inside the embryo.


Subject(s)
Apoptosis , Cell Lineage , DNA Methylation , Endoderm , Gene Expression Regulation, Developmental , Animals , Endoderm/cytology , Endoderm/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Phagocytosis , Mice, Inbred C57BL , Mice , Cell Differentiation , Female , Embryonic Development , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Mice, Transgenic , Gastrointestinal Tract/cytology , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism
8.
Nat Commun ; 15(1): 5247, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898078

ABSTRACT

DNA replication in differentiated cells follows a defined program, but when and how it is established during mammalian development is not known. Here we show using single-cell sequencing, that late replicating regions are established in association with the B compartment and the nuclear lamina from the first cell cycle after fertilization on both maternal and paternal genomes. Late replicating regions contain a relative paucity of active origins and few but long genes and low G/C content. In both bovine and mouse embryos, replication timing patterns are established prior to embryonic genome activation. Chromosome breaks, which form spontaneously in bovine embryos at sites concordant with human embryos, preferentially locate to late replicating regions. In mice, late replicating regions show enhanced fragility due to a sparsity of dormant origins that can be activated under conditions of replication stress. This pattern predisposes regions with long neuronal genes to fragility and genetic change prior to separation of soma and germ cell lineages. Our studies show that the segregation of early and late replicating regions is among the first layers of genome organization established after fertilization.


Subject(s)
DNA Replication , Embryo, Mammalian , Nuclear Lamina , Animals , Mice , Embryo, Mammalian/metabolism , Cattle , Nuclear Lamina/metabolism , Female , Male , Humans , Embryonic Development/genetics , Genome , Single-Cell Analysis
9.
Nature ; 630(8017): 720-727, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839949

ABSTRACT

Spermatozoa harbour a complex and environment-sensitive pool of small non-coding RNAs (sncRNAs)1, which influences offspring development and adult phenotypes1-7. Whether spermatozoa in the epididymis are directly susceptible to environmental cues is not fully understood8. Here we used two distinct paradigms of preconception acute high-fat diet to dissect epididymal versus testicular contributions to the sperm sncRNA pool and offspring health. We show that epididymal spermatozoa, but not developing germ cells, are sensitive to the environment and identify mitochondrial tRNAs (mt-tRNAs) and their fragments (mt-tsRNAs) as sperm-borne factors. In humans, mt-tsRNAs in spermatozoa correlate with body mass index, and paternal overweight at conception doubles offspring obesity risk and compromises metabolic health. Sperm sncRNA sequencing of mice mutant for genes involved in mitochondrial function, and metabolic phenotyping of their wild-type offspring, suggest that the upregulation of mt-tsRNAs is downstream of mitochondrial dysfunction. Single-embryo transcriptomics of genetically hybrid two-cell embryos demonstrated sperm-to-oocyte transfer of mt-tRNAs at fertilization and suggested their involvement in the control of early-embryo transcription. Our study supports the importance of paternal health at conception for offspring metabolism, shows that mt-tRNAs are diet-induced and sperm-borne and demonstrates, in a physiological setting, father-to-offspring transfer of sperm mitochondrial RNAs at fertilization.


Subject(s)
Diet, High-Fat , Epigenesis, Genetic , Mitochondria , RNA, Mitochondrial , Spermatozoa , Animals , Female , Humans , Male , Mice , Body Mass Index , Diet, High-Fat/adverse effects , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Epididymis/cytology , Epigenesis, Genetic/genetics , Fertilization/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Obesity/genetics , Obesity/metabolism , Obesity/etiology , Oocytes/metabolism , Overweight/genetics , Overweight/metabolism , Paternal Inheritance/genetics , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Spermatozoa/metabolism , Testis/cytology , Transcription, Genetic
10.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892059

ABSTRACT

Global methylation levels differ in in vitro- and in vivo-developed embryos. Follicular fluid (FF) contains extracellular vesicles (EVs) containing miRNAs that affect embryonic development. Here, we examined our hypothesis that components in FF affect global DNA methylation and embryonic development. Oocytes and FF were collected from bovine ovaries. Treatment of zygotes with a low concentration of FF induced global DNA demethylation, improved embryonic development, and reduced DNMT1/3A levels. We show that embryos take up EVs containing labeled miRNA secreted from granulosa cells and the treatment of zygotes with EVs derived from FF reduces global DNA methylation in embryos. Furthermore, the methylation levels of in vitro-developed blastocysts were higher than those of in their vivo counterparts. Based on small RNA-sequencing and in silico analysis, we predicted miR-29b, -199a-3p, and -148a to target DNMTs and to induce DNA demethylation, thereby improving embryonic development. Moreover, among FF from 30 cows, FF with a high content of these miRNAs demethylated more DNA in the embryos than FF with a lower miRNA content. Thus, miRNAs in FF play a role in early embryonic development.


Subject(s)
Embryonic Development , Extracellular Vesicles , Follicular Fluid , MicroRNAs , Animals , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Cattle , Follicular Fluid/metabolism , Extracellular Vesicles/metabolism , Embryonic Development/genetics , DNA Methylation , DNA Demethylation , Oocytes/metabolism , Blastocyst/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Zygote/metabolism
11.
Dev Cell ; 59(12): 1487-1488, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38889690

ABSTRACT

In this issue of Developmental Cell, Bolondi et al. systematically assesses neuro-mesodermal progenitor (NMP) dynamics by combining a mouse stem-cell-based embryo model with molecular recording of single cells, shedding light on the dynamics of neural tube and paraxial mesoderm formation during mammalian development.


Subject(s)
Mesoderm , Animals , Mice , Mesoderm/cytology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Neural Tube/cytology , Neural Tube/embryology , Cell Differentiation/physiology , Stem Cells/cytology , Stem Cells/metabolism , Body Patterning
12.
Reprod Fertil Dev ; 362024 Jun.
Article in English | MEDLINE | ID: mdl-38902907

ABSTRACT

Context Current methods to obtain bovine embryos of high genetic merit include approaches that require skilled techniques for low-efficiency cloning strategies. Aims The overall goal herein was to identify the efficacy of alternative methods for producing multiple embryos through blastomere complementation while determining maintenance of cell pluripotency. Methods Bovine oocytes were fertilised in vitro to produce 4-cell embryos from which blastomeres were isolated and cultured as 2-cell aggregates using a well-of-the-well system. Aggregates were returned to incubation up to 7days (Passage 1). A second passage of complement embryos was achieved by splitting 4-cell Passage 1 embryos. Passaged embryos reaching the blastocyst stage were characterised for cell number and cell lineage specification in replicate with non-reconstructed zona-intact embryos. Key results Passage 1 and 2 embryo complements yielded 29% and 25% blastocyst development, respectively. Passage 1 embryos formed blastocysts, but with a reduction in expression of SOX2 and decreased size compared to non-reconstructed zona-intact embryos. Passage 2 embryos had a complete lack of SOX2 expression and a reduction in transcript abundance of SOX2 and SOX17, suggesting loss of pluripotency markers that primarily affected inner cell mass (ICM) and hypoblast formation. Conclusions In vitro fertilised bovine embryos can be reconstructed with multiple passaging to generate genetically identical embryos. Increased passaging drives trophectoderm cell lineage specification while compromising ICM formation. Implications These results may provide an alternative strategy for producing genetically identical bovine embryos through blastomere complementation with applications towards the development of trophoblast and placental models of early development.


Subject(s)
Blastocyst , Blastomeres , Embryo Culture Techniques , Embryonic Development , Fertilization in Vitro , Animals , Cattle , Blastocyst/metabolism , Fertilization in Vitro/veterinary , Embryo Culture Techniques/veterinary , Embryonic Development/physiology , Blastomeres/metabolism , Blastomeres/cytology , Female , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cloning, Organism/methods , Cloning, Organism/veterinary , Cell Lineage , Embryo, Mammalian/metabolism
13.
Cells ; 13(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38891086

ABSTRACT

Reduced oxygen during embryo culture in human ART prevents embryo oxidative stress. Oxidative stress is also the major mechanism by which maternal diabetes impairs embryonic development. This study employed induced hyperglycemia prepubertal mice to mimic childhood diabetes to understand the effects of varying oxygen tension during in vitro embryonic development. The oocytes were fertilized and cultured at low (≈5%) oxygen (LOT) or atmospheric (≈20%) oxygen tension (HOT) for up to 96 h. Embryo development, apoptosis in blastocysts, inner cell mass (ICM) outgrowth proliferation, and Hif1α expression were assessed. Though the oocyte quality and meiotic spindle were not affected, the fertilization rate (94.86 ± 1.18 vs. 85.17 ± 2.81), blastocyst rate (80.92 ± 2.92 vs. 69.32 ± 2.54), and ICM proliferation ability (51.04 ± 9.22 vs. 17.08 ± 3.05) of the hyperglycemic embryos were significantly higher in the LOT compared to the HOT group. On the other hand, blastocysts from the hyperglycemic group, cultured at HOT, had a 1.5-fold increase in apoptotic cells compared to the control and lower Hif1α transcripts in ICM outgrowths compared to the LOT. Increased susceptibility of embryos from hyperglycemic mice to higher oxygen tension warrants the need to individualize the conditions for embryo culture systems in ART clinics, particularly when an endogenous maternal pathology affects the ovarian environment.


Subject(s)
Embryonic Development , Hyperglycemia , Oxygen , Animals , Female , Oxygen/metabolism , Oxygen/pharmacology , Mice , Hyperglycemia/metabolism , Hyperglycemia/pathology , Embryonic Development/genetics , Apoptosis/drug effects , Blastocyst/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Oocytes/metabolism , Embryo, Mammalian/metabolism , Cell Proliferation
14.
Nat Commun ; 15(1): 5210, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890321

ABSTRACT

Cell-fate decisions during mammalian gastrulation are poorly understood outside of rodent embryos. The embryonic disc of pig embryos mirrors humans, making them a useful proxy for studying gastrulation. Here we present a single-cell transcriptomic atlas of pig gastrulation, revealing cell-fate emergence dynamics, as well as conserved and divergent gene programs governing early porcine, primate, and murine development. We highlight heterochronicity in extraembryonic cell-types, despite the broad conservation of cell-type-specific transcriptional programs. We apply these findings in combination with functional investigations, to outline conserved spatial, molecular, and temporal events during definitive endoderm specification. We find early FOXA2 + /TBXT- embryonic disc cells directly form definitive endoderm, contrasting later-emerging FOXA2/TBXT+ node/notochord progenitors. Unlike mesoderm, none of these progenitors undergo epithelial-to-mesenchymal transition. Endoderm/Node fate hinges on balanced WNT and hypoblast-derived NODAL, which is extinguished upon endodermal differentiation. These findings emphasise the interplay between temporal and topological signalling in fate determination during gastrulation.


Subject(s)
Embryo, Mammalian , Endoderm , Gastrulation , Gene Expression Regulation, Developmental , Single-Cell Analysis , Animals , Endoderm/cytology , Endoderm/metabolism , Endoderm/embryology , Swine , Mice , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Cell Differentiation , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Transcriptome , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Cell Lineage , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Epithelial-Mesenchymal Transition/genetics
15.
FASEB J ; 38(11): e23738, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38855924

ABSTRACT

Maternal nutrition contributes to gene-environment interactions that influence susceptibility to common congenital anomalies such as neural tube defects (NTDs). Supplemental myo-inositol (MI) can prevent NTDs in some mouse models and shows potential for prevention of human NTDs. We investigated effects of maternal MI intake on embryonic MI status and metabolism in curly tail mice, which are genetically predisposed to NTDs that are inositol-responsive but folic acid resistant. Dietary MI deficiency caused diminished MI in maternal plasma and embryos, showing that de novo synthesis is insufficient to maintain MI levels in either adult or embryonic mice. Under normal maternal dietary conditions, curly tail embryos that developed cranial NTDs had significantly lower MI content than unaffected embryos, revealing an association between diminished MI status and failure of cranial neurulation. Expression of inositol-3-phosphate synthase 1, required for inositol biosynthesis, was less abundant in the cranial neural tube than at other axial levels. Supplemental MI or d-chiro-inositol (DCI) have previously been found to prevent NTDs in curly tail embryos. Here, we investigated the metabolic effects of MI and DCI treatments by mass spectrometry-based metabolome analysis. Among inositol-responsive metabolites, we noted a disproportionate effect on nucleotides, especially purines. We also found altered proportions of 5-methyltetrahydrolate and tetrahydrofolate in MI-treated embryos suggesting altered folate metabolism. Treatment with nucleotides or the one-carbon donor formate has also been found to prevent NTDs in curly tail embryos. Together, these findings suggest that the protective effect of inositol may be mediated through the enhanced supply of nucleotides during neural tube closure.


Subject(s)
Inositol , Neural Tube Defects , Inositol/metabolism , Inositol/pharmacology , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , Animals , Female , Mice , Pregnancy , Embryo, Mammalian/metabolism , Maternal Nutritional Physiological Phenomena , Metabolome , Folic Acid/metabolism
16.
Curr Top Dev Biol ; 160: 31-64, 2024.
Article in English | MEDLINE | ID: mdl-38937030

ABSTRACT

Biomechanics in embryogenesis is a dynamic field intertwining the physical forces and biological processes that shape the first days of a mammalian embryo. From the first cell fate bifurcation during blastulation to the complex symmetry breaking and tissue remodeling in gastrulation, mechanical cues appear critical in cell fate decisions and tissue patterning. Recent strides in mouse and human embryo culture, stem cell modeling of mammalian embryos, and biomaterial design have shed light on the role of cellular forces, cell polarization, and the extracellular matrix in influencing cell differentiation and morphogenesis. This chapter highlights the essential functions of biophysical mechanisms in blastocyst formation, embryo implantation, and early gastrulation where the interplay between the cytoskeleton and extracellular matrix stiffness orchestrates the intricacies of embryogenesis and placenta specification. The advancement of in vitro models like blastoids, gastruloids, and other types of embryoids, has begun to faithfully recapitulate human development stages, offering new avenues for exploring the biophysical underpinnings of early development. The integration of synthetic biology and advanced biomaterials is enhancing the precision with which we can mimic and study these processes. Looking ahead, we emphasize the potential of CRISPR-mediated genomic perturbations coupled with live imaging to uncover new mechanosensitive pathways and the application of engineered biomaterials to fine-tune the mechanical conditions conducive to embryonic development. This synthesis not only bridges the gap between experimental models and in vivo conditions to advancing fundamental developmental biology of mammalian embryogenesis, but also sets the stage for leveraging biomechanical insights to inform regenerative medicine.


Subject(s)
Embryonic Development , Animals , Humans , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Biomechanical Phenomena
17.
Cell Stress Chaperones ; 29(3): 497-509, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38763404

ABSTRACT

Bcl2-associated athanogene-1 protein (Bag1) acts as a co-chaperone of heat shock protein 70 and heat shock cognate 70 and regulates multiple cellular processes, including cell proliferation, apoptosis, environmental stress response, and drug resistance. Since Bag1 knockout mice exhibited fetal lethality, the in vivo function of Bag1 remains unclear. In this study, we established a mouse line expressing Bag1 gene missing exon 5, which corresponds to an encoding region for the interface of heat shock protein 70/heat shock cognate 70. Despite mice carrying homoalleles of the Bag1 mutant (Bag1Δex5) expressing undetectable levels of Bag1, Bag1Δex5 homozygous mice developed without abnormalities. Bag1Δex5 protein was found to be highly unstable in cells and in vitro. We found that the growth of mouse embryonic fibroblasts derived from Bag1Δex5-homo mice was attenuated by doxorubicin and a glutathione (GSH) synthesis inhibitor, buthionine sulfoximine. In response to buthionine sulfoximine, Bag1Δex5-mouse embryonic fibroblasts exhibited a higher dropping rate of GSH relative to the oxidized glutathione level. In addition, Bag1 might mitigate cellular hydrogen peroxide levels. Taken together, our results demonstrate that the loss of Bag1 did not affect mouse development and that Bag1 is involved in intracellular GSH homeostasis, namely redox homeostasis.


Subject(s)
DNA-Binding Proteins , Fibroblasts , Glutathione , Transcription Factors , Animals , Fibroblasts/metabolism , Glutathione/metabolism , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Doxorubicin/pharmacology , Buthionine Sulfoximine/pharmacology , Embryo, Mammalian/metabolism , Cell Proliferation , Mice, Knockout , Hydrogen Peroxide/metabolism
18.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38691188

ABSTRACT

Analysis of single cell transcriptomics (scRNA-seq) data is typically performed after subsetting to highly variable genes (HVGs). Here, we show that Entropy Sorting provides an alternative mathematical framework for feature selection. On synthetic datasets, continuous Entropy Sort Feature Weighting (cESFW) outperforms HVG selection in distinguishing cell-state-specific genes. We apply cESFW to six merged scRNA-seq datasets spanning human early embryo development. Without smoothing or augmenting the raw counts matrices, cESFW generates a high-resolution embedding displaying coherent developmental progression from eight-cell to post-implantation stages and delineating 15 distinct cell states. The embedding highlights sequential lineage decisions during blastocyst development, while unsupervised clustering identifies branch point populations obscured in previous analyses. The first branching region, where morula cells become specified for inner cell mass or trophectoderm, includes cells previously asserted to lack a developmental trajectory. We quantify the relatedness of different pluripotent stem cell cultures to distinct embryo cell types and identify marker genes of naïve and primed pluripotency. Finally, by revealing genes with dynamic lineage-specific expression, we provide markers for staging progression from morula to blastocyst.


Subject(s)
Cell Lineage , Embryo, Mammalian , Embryonic Development , Entropy , Single-Cell Analysis , Transcriptome , Humans , Transcriptome/genetics , Single-Cell Analysis/methods , Embryonic Development/genetics , Embryo, Mammalian/metabolism , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Blastocyst/metabolism , Blastocyst/cytology , Gene Expression Profiling , Morula/metabolism , Morula/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology
19.
Dis Model Mech ; 17(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38804708

ABSTRACT

The TATA box-binding protein-associated factor 1 (TAF1) is a ubiquitously expressed protein and the largest subunit of the basal transcription factor TFIID, which plays a key role in initiation of RNA polymerase II-dependent transcription. TAF1 missense variants in human males cause X-linked intellectual disability, a neurodevelopmental disorder, and TAF1 is dysregulated in X-linked dystonia-parkinsonism, a neurodegenerative disorder. However, this field has lacked a genetic mouse model of TAF1 disease to explore its mechanism in mammals and treatments. Here, we generated and validated a conditional cre-lox allele and the first ubiquitous Taf1 knockout mouse. We discovered that Taf1 deletion in male mice was embryonically lethal, which may explain why no null variants have been identified in humans. In the brains of Taf1 heterozygous female mice, no differences were found in gross structure, overall expression and protein localisation, suggesting extreme skewed X inactivation towards the non-mutant chromosome. Nevertheless, these female mice exhibited a significant increase in weight, weight with age, and reduced movement, suggesting that a small subset of neurons was negatively impacted by Taf1 loss. Finally, this new mouse model may be a future platform for the development of TAF1 disease therapeutics.


Subject(s)
Body Weight , Heterozygote , Histone Acetyltransferases , Mice, Knockout , Movement Disorders , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Animals , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/deficiency , Female , Male , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Movement Disorders/genetics , Movement Disorders/pathology , Embryo, Mammalian/metabolism , Mice , Brain/pathology , Brain/metabolism , Genes, Lethal , Mice, Inbred C57BL
20.
Stem Cell Reports ; 19(6): 906-921, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38729154

ABSTRACT

Removal of somatic histone H3 lysine 9 trimethylation (H3K9me3) from the embryonic genome can improve the efficiency of mammalian cloning using somatic cell nuclear transfer (SCNT). However, this strategy involves the injection of histone demethylase mRNA into embryos, which is limiting because of its invasive and labor-consuming nature. Here, we report that treatment with an inhibitor of G9a (G9ai), the major histone methyltransferase that introduces H3K9me1/2 in mammals, greatly improved the development of mouse SCNT embryos. Intriguingly, G9ai caused an immediate reduction of H3K9me1/2, a secondary loss of H3K9me3 in SCNT embryos, and increased the birth rate of cloned pups about 5-fold (up to 3.9%). G9ai combined with the histone deacetylase inhibitor trichostatin A further improved this rate to 14.5%. Mechanistically, G9ai and TSA synergistically enhanced H3K9me3 demethylation and boosted zygotic genome activation. Thus, we established an easy, highly effective SCNT protocol that would enhance future cloning research and applications.


Subject(s)
Histone-Lysine N-Methyltransferase , Histones , Nuclear Transfer Techniques , Animals , Histones/metabolism , Mice , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Methylation , Cloning, Organism/methods , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Embryonic Development/genetics , Hydroxamic Acids/pharmacology , Female , Histone Deacetylase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...