Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Proc Natl Acad Sci U S A ; 120(52): e2313200120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38113263

ABSTRACT

In female mice, the gene dosage from X chromosomes is adjusted by a process called X chromosome inactivation (XCI) that occurs in two steps. An imprinted form of XCI (iXCI) that silences the paternally inherited X chromosome (Xp) is initiated at the 2- to 4-cell stages. As extraembryonic cells including trophoblasts keep the Xp silenced, epiblast cells that give rise to the embryo proper reactivate the Xp and undergo a random form of XCI (rXCI) around implantation. Both iXCI and rXCI require the lncRNA Xist, which is expressed from the X to be inactivated. The X-linked E3 ubiquitin ligase Rlim (Rnf12) in conjunction with its target protein Rex1 (Zfp42), a critical repressor of Xist, have emerged as major regulators of iXCI. However, their roles in rXCI remain controversial. Investigating early mouse development, we show that the Rlim-Rex1 axis is active in pre-implantation embryos. Upon implantation Rex1 levels are downregulated independently of Rlim specifically in epiblast cells. These results provide a conceptual framework of how the functional dynamics between Rlim and Rex1 ensures regulation of iXCI but not rXCI in female mice.


Subject(s)
RNA, Long Noncoding , X Chromosome Inactivation , Animals , Female , Mice , Embryo, Mammalian/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , X Chromosome/genetics , X Chromosome/metabolism , X Chromosome Inactivation/genetics
2.
Dev Biol ; 511: 53-62, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38593904

ABSTRACT

Early embryonic development is a finely orchestrated process that requires precise regulation of gene expression coordinated with morphogenetic events. TATA-box binding protein-associated factors (TAFs), integral components of transcription initiation coactivators like TFIID and SAGA, play a crucial role in this intricate process. Here we show that disruptions in TAF5, TAF12 and TAF13 individually lead to embryonic lethality in the mouse, resulting in overlapping yet distinct phenotypes. Taf5 and Taf12 mutant embryos exhibited a failure to implant post-blastocyst formation, and Taf5 mutants have aberrant lineage specification within the inner cell mass. In contrast, Taf13 mutant embryos successfully implant and form egg-cylinder stages but fail to initiate gastrulation. Strikingly, we observed a depletion of pluripotency factors in TAF13-deficient embryos, including OCT4, NANOG and SOX2, highlighting an indispensable role of TAF13 in maintaining pluripotency. Transcriptomic analysis revealed distinct gene targets affected by the loss of TAF5, TAF12 and TAF13. Thus, we propose that TAF5, TAF12 and TAF13 convey locus specificity to the TFIID complex throughout the mouse genome.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , TATA-Binding Protein Associated Factors , Animals , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/genetics , Mice , Embryonic Development/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/genetics , Female , Blastocyst/metabolism , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Gastrulation/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Embryo, Mammalian/metabolism
3.
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
4.
Dev Biol ; 491: 31-42, 2022 11.
Article in English | MEDLINE | ID: mdl-36028102

ABSTRACT

Retinoic acid (RA), a metabolite of vitamin A, is a small molecule and morphogen that is required for embryonic development. While normal RA signals are required for hepatic development in a variety of vertebrates, a role for RA during mammalian hepatic specification has yet to be defined. To examine the requirement for RA in murine liver induction, we performed whole embryo culture with the small molecule RA inhibitor, BMS493, to attenuate RA signaling immediately prior to hepatic induction and through liver bud formation. BMS493 treated embryos demonstrated a significant loss of hepatic specification that was confined to the prospective dorsal anterior liver bud. Examination of RA attenuated embryos demonstrates that while the liver bud displays normal expression of foregut endoderm markers and the hepato-pancreatobiliary domain marker, PROX1, the dorsal/anterior liver bud excludes the critical hepatic marker, HNF4α, indicating that RA signals are required for dorsal/anterior hepatic induction. These results were confirmed and extended by careful examination of Rdh10<sup>trex/trex</sup> embryos, which carry a genetic perturbation in RA synthesis. At E9.5 Rdh10<sup>trex/trex</sup> embryos display a similar yet more significant loss of the anterior/dorsal liver bud. Notably the anterior/dorsal liver bud loss correlates with the known dorsal-ventral gradient of the RA synthesis enzyme, Aldh1a2. In addition to altered hepatic specification, the mesoderm surrounding the liver bud is disorganized in RA abrogated embryos. Analysis of E10.5 Rdh10<sup>trex/trex</sup> embryos reveals small livers that appear to lack the dorsal/caudal lobes. Finally, addition of exogenous RA prior to hepatic induction results in a liver bud that has failed to thicken and is largely unspecified. Taken together our ex vivo and in vivo evidence demonstrate that the generation of normal RA gradients is required for hepatic patterning, specification, and growth.


Subject(s)
Tretinoin , Vitamin A , Animals , Endoderm/metabolism , Female , Liver , Mammals/metabolism , Mice , Pregnancy , Prospective Studies , Tretinoin/metabolism , Tretinoin/pharmacology , Vitamin A/metabolism
5.
Development ; 147(10)2020 05 26.
Article in English | MEDLINE | ID: mdl-32376682

ABSTRACT

Mitochondria are essential for energy production and although they have their own genome, many nuclear-encoded mitochondrial ribosomal proteins (MRPs) are required for proper function of the organelle. Although mutations in MRPs have been associated with human diseases, little is known about their role during development. Presented here are the null phenotypes for 21 nuclear-encoded mitochondrial proteins and in-depth characterization of mouse embryos mutant for the Mrp genes Mrpl3, Mrpl22, Mrpl44, Mrps18c and Mrps22 Loss of each MRP results in successful implantation and egg-cylinder formation, followed by severe developmental delay and failure to initiate gastrulation by embryonic day 7.5. The robust and similar single knockout phenotypes are somewhat surprising given there are over 70 MRPs and suggest little functional redundancy. Metabolic analysis reveals that Mrp knockout embryos produce significantly less ATP than controls, indicating compromised mitochondrial function. Histological and immunofluorescence analyses indicate abnormal organelle morphology and stalling at the G2/M checkpoint in Mrp null cells. The nearly identical pre-gastrulation phenotype observed for many different nuclear-encoded mitochondrial protein knockouts hints that distinct energy systems are crucial at specific time points during mammalian development.


Subject(s)
Embryonic Development/genetics , Gastrulation/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Ribosomes/metabolism , Ribosomal Proteins/genetics , Animals , Cell Cycle Checkpoints/genetics , Female , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
6.
Mol Reprod Dev ; 90(2): 98-108, 2023 02.
Article in English | MEDLINE | ID: mdl-36528861

ABSTRACT

As a highly conserved DNA polymerase (Pol), Pol δ plays crucial roles in chromosomal DNA synthesis and various DNA repair pathways. However, the function of POLD2, the second small subunit of DNA Pol δ (p50 subunit), has not been characterized in vivo during mammalian development. Here, we report for the first time, the essential role of subunit POLD2 during early murine embryogenesis. Although Pold2 mutant mouse embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at gastrulation stages. Outgrowth assays reveal that mutant blastocysts cannot hatch from the zona pellucida, indicating impaired blastocyst function. Notably, these phenotypes can be recapitulated by small interfering RNA (siRNA)-mediated knockdown, which also exhibit slowed cellular proliferation together with skewed primitive endoderm and epiblast allocation during the second cell lineage specification. In summary, our study demonstrates that POLD2 is essential for the earliest steps of mammalian development, and the retarded proliferation and embryogenesis may also alter the following cell lineage specifications in the mouse blastocyst embryos.


Subject(s)
Blastocyst , DNA Polymerase III , Embryonic Development , Animals , Mice , Blastocyst/metabolism , Cell Lineage , Endoderm/metabolism , Germ Layers , Mammals , DNA Polymerase III/metabolism
7.
Mol Reprod Dev ; 89(8): 337-350, 2022 08.
Article in English | MEDLINE | ID: mdl-35726782

ABSTRACT

Early development and differentiation require precise control of cellular functions. Lysosomal degradation is a critical component of normal cellular homeostasis, allowing for degradation of signaling molecules, proteins, and other macromolecules for cellular remodeling and signaling. Little is known about the role of lysosomal function in mammalian embryos before gastrulation. Borcs6 is a protein involved in lysosomal trafficking as well as endo-lysosomal and autophagosome fusion. Here, we show that Borcs6 is necessary for efficient endo-lysosomal degradation in the early embryo. Although embryos lacking Borcs6 are developmentally comparable to control littermates at E5.5, they are characterized by large cells containing increased levels of late endosomes and abnormal nuclei. Furthermore, these embryos display a skewed ratio of extraembryonic and embryonic cell lineages, are delayed by E6.5, and do not undergo normal gastrulation. These results demonstrate the essential functions of lysosomal positioning and fusion with endosomes during early embryonic development and indicate that the early lethality of BORCS6 mutant embryos is primarily due to defects in the HOPS-related function of BORC rather than lysosomal positioning.


Subject(s)
Endosomes , Lysosomes , Animals , Autophagy , Endosomes/metabolism , Homeostasis , Lysosomes/metabolism , Mammals , Membrane Fusion , Proteins/metabolism
8.
Dev Biol ; 465(1): 1-10, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32628936

ABSTRACT

Protein phosphatases regulate a wide array of proteins through post-translational modification and are required for a plethora of intracellular events in eukaryotes. While some core components of the protein phosphatase complexes are well characterized, many subunits of these large complexes remain unstudied. Here we characterize a loss-of-function allele of the protein phosphatase 1 regulatory subunit 35 (Ppp1r35) gene. Homozygous mouse embryos lacking Ppp1r35 are developmental delayed beginning at embryonic day (E) 7.5 and have obvious morphological defects at later stages. Mutants fail to initiate turning and do not progress beyond the size or staging of normal E8.5 embryos. Consistent with recent in vitro studies linking PPP1R35 with the microcephaly protein Rotatin and with a role in centrosome formation, we show that Ppp1r35 mutant embryos lack primary cilia. Histological and molecular analysis of Ppp1r35 mutants revealed that notochord development is irregular and discontinuous and consistent with a role in primary cilia, that the floor plate of the neural tube is not specified. Similar to other mutant embryos with defects in centriole function, Ppp1r35 mutants displayed increased cell death that is prevalent in the neural tube and an increased number of proliferative cells in prometaphase. We hypothesize that loss of Ppp1r35 function abrogates centriole homeostasis, resulting in a failure to produce functional primary cilia, cell death and cell cycle delay/stalling that leads to developmental failure. Taken together, these results highlight the essential function of Ppp1r35 during early mammalian development and implicate this gene as a candidate for human microcephaly.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , Cilia/metabolism , Notochord/enzymology , Organogenesis , Animals , Cell Cycle Proteins/genetics , Cilia/genetics , Mice , Mice, Knockout
9.
Hum Mol Genet ; 28(16): 2775-2784, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31107948

ABSTRACT

The dynein axonemal assembly factor (Dnaaf) protein family is involved in preassembly and stability of dynein arms before they are transported into the cilia. In humans, mutations in DNAAF genes lead to several diseases related to cilia defects such as primary ciliary dyskinesia (PCD; OMIM: 612518). Patients with PCD experience malfunctions in cilia motility, which can result in inflammation and infection of the respiratory tract among other defects. Previous studies have identified that a mutation in DNAAF2 results in PCD and that 40% of these patients also experience laterality defects. In an outbred genetic background, Dnaaf2 homozygotes die after birth and have left/right defects among other phenotypes. Here we characterize a novel null allele of Dnaaf2 obtained from the International Mouse Phenotyping Consortium. Our data indicate that on a defined C57bl/6NJ genetic background, homozygous Dnaaf2 mouse embryos fail to progress beyond organogenesis stages with many abnormalities including left-right patterning defects. These findings support studies indicating that hypomorphic mutations of human DNAAF2 can result in ciliary dyskinesia and identify Dnaaf2 as an essential component of cilia function in vivo.


Subject(s)
Ciliary Motility Disorders/etiology , Genes, Lethal , Microtubule-Associated Proteins/deficiency , Mutation , Alleles , Animals , Body Patterning , Ciliary Motility Disorders/metabolism , Disease Models, Animal , Embryonic Development/genetics , Gene Expression , Genotype , Mice , Phenotype
10.
Development ; 145(19)2018 10 11.
Article in English | MEDLINE | ID: mdl-30232173

ABSTRACT

The definitive endoderm (DE) is the embryonic germ layer that forms the gut tube and associated organs, including thymus, lungs, liver and pancreas. To understand how individual DE cells furnish gut organs, genetic fate mapping was performed using the Rosa26lacZ Cre-reporter paired with a tamoxifen-inducible DE-specific Cre-expressing transgene. We established a low tamoxifen dose that infrequently induced heritable lacZ expression in a single cell of individual E8.5 mouse embryos and identified clonal cell descendants at E16.5. As expected, only a fraction of the E16.5 embryos contained lacZ-positive clonal descendants and a subset of these contained descendants in multiple organs, revealing novel ontogeny. Furthermore, immunohistochemical analysis was used to identify lacZ-positive hepatocytes and biliary epithelial cells, which are the cholangiocyte precursors, in each clonally populated liver. Together, these data not only uncover novel and suspected lineage relationships between DE-derived organs, but also illustrate the bipotential nature of individual hepatoblasts by demonstrating that single hepatoblasts contribute to both the hepatocyte and the cholangiocyte lineage in vivo.


Subject(s)
Chromosome Mapping , Endoderm/cytology , Hepatocytes/cytology , Organ Specificity/genetics , Single-Cell Analysis , Stem Cells/cytology , Animals , Bile Ducts/cytology , Cell Lineage , Clone Cells , Embryo, Mammalian/cytology , Epithelial Cells/cytology , Female , Hepatocytes/metabolism , Male , Mice, Inbred C57BL , Stem Cells/metabolism
11.
Biol Reprod ; 104(2): 325-335, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33246328

ABSTRACT

Zinc finger domains of the Cys-Cys-Cys-His (CCCH) class are evolutionarily conserved proteins that bind nucleic acids and are involved in various biological processes. Nearly 60 CCCH-type zinc finger proteins have been identified in humans and mice, most have not been functionally characterized. Here, we provide the first in vivo functional characterization of ZC3H4-a novel CCCH-type zinc finger protein. Our results show that although Zc3h4 mutant embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at E7.5 early post-gastrulation stage, suggesting implantation failure. Outgrowth assays reveal that mutant blastocysts either fail to hatch from the zona pellucida, or can hatch but do not form a typical inner cell mass colony, the source of embryonic stem cells (ESCs). Although there is no change in levels of reactive oxygen species, Zc3h4 mutants display severe DNA breaks and reduced cell proliferation. Analysis of lineage specification reveals that both epiblast and primitive endoderm lineages are compromised with severe reductions in cell number and/or specification in the mutant blastocysts. In summary, these findings demonstrate the essential role of ZC3H4 during early mammalian embryogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo Implantation/physiology , Embryonic Development/physiology , Animals , Cell Proliferation/genetics , DNA Breaks , DNA-Binding Proteins/genetics , Embryo Implantation/genetics , Female , Gene Expression Regulation, Developmental , Genotype , Mice , Mice, Knockout , Mutation
12.
Biol Reprod ; 103(1): 13-23, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32285100

ABSTRACT

Retinoblastoma-binding protein 4 (RBBP4) (also known as chromatin-remodeling factor RBAP48) is an evolutionarily conserved protein that has been involved in various biological processes. Although a variety of functions have been attributed to RBBP4 in vitro, mammalian RBBP4 has not been studied in vivo. Here we report that RBBP4 is essential during early mouse embryo development. Although Rbbp4 mutant embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at E7.5 early post-gastrulation stage, suggesting an implantation failure. Outgrowth (OG) assays reveal that mutant blastocysts cannot hatch from the zona or can hatch but then arrest without further development. We find that while there is no change in proliferation or levels of reactive oxygen species, both apoptosis and histone acetylation are significantly increased in mutant blastocysts. Analysis of lineage specification reveals that while the trophoblast is properly specified, both epiblast and primitive endoderm lineages are compromised with severe reductions in cell number and/or specification. In summary, these findings demonstrate the essential role of RBBP4 during early mammalian embryogenesis.


Subject(s)
Apoptosis , Blastocyst/physiology , Embryo Loss , Endoderm/embryology , Histones/metabolism , Retinoblastoma-Binding Protein 4/physiology , Acetylation , Animals , Embryo Implantation/physiology , Embryonic Development/physiology , Endoderm/cytology , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinoblastoma-Binding Protein 4/deficiency , Retinoblastoma-Binding Protein 4/genetics
13.
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
14.
Mol Reprod Dev ; 87(11): 1152-1158, 2020 11.
Article in English | MEDLINE | ID: mdl-33022126

ABSTRACT

In eukaryotic cells, RNA polymerase (Pol) I and Pol III are dedicated to the synthesis of ribosomal RNA precursors and a variety of small RNAs, respectively. Although RNA Pol I and Pol III complexes are crucial for the regulation of cell growth and cell cycle in all cell types, many of the components of the Pol I and Pol III complexes have not been functionally characterized in mammals. Here, we provide the first in vivo functional characterization of POLR1D, a subunit shared by RNA Pol I and Pol III, during early mammalian embryo development. Our results show that Polr1d mutant embryos cannot be recovered at E7.5 early post-gastrulation stage, suggesting failed implantation. Although Polr1d mutants can be recovered at E3.5, they exhibit delayed/stalled development with morula morphology rather than differentiation into blastocysts. Even with extended time in culture, mutant embryos fail to form blastocysts and eventually die. Analysis of E3.0 embryos revealed severe DNA damage in Polr1d mutants. Additionally, lineage assessment reveals that trophectoderm specification is compromised in the absence of Polr1d. In summary, these findings demonstrate the essential role of POLR1D during early mammalian embryogenesis and highlight cell-lethal phenotype without Polr1d function.


Subject(s)
DNA-Directed RNA Polymerases/deficiency , Embryo, Mammalian/metabolism , Embryonic Development , Animals , Blastocyst , CRISPR-Cas Systems , DNA Damage , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/physiology , Exons/genetics , Female , Gastrulation , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Genes, Lethal , Gestational Age , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Morula/chemistry , Morula/ultrastructure , Organ Culture Techniques , Organelle Biogenesis , Pregnancy , Reactive Oxygen Species/analysis , Ribosomes , Sequence Deletion
15.
Nature ; 511(7507): 86-9, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24870238

ABSTRACT

In female mice, two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Beginning at the four-cell stage, imprinted XCI (iXCI) exclusively silences the paternal X chromosome. Later, around implantation, epiblast cells of the inner cell mass that give rise to the embryo reactivate the paternal X chromosome and undergo a random form of XCI (rXCI). Xist, a long non-coding RNA crucial for both forms of XCI, is activated by the ubiquitin ligase RLIM (also known as Rnf12). Although RLIM is required for triggering iXCI in mice, its importance for rXCI has been controversial. Here we show that RLIM levels are downregulated in embryonic cells undergoing rXCI. Using mouse genetics we demonstrate that female cells lacking RLIM from pre-implantation stages onwards show hallmarks of XCI, including Xist clouds and H3K27me3 foci, and have full embryogenic potential. These results provide evidence that RLIM is dispensable for rXCI, indicating that in mice an RLIM-independent mechanism activates Xist in the embryo proper.


Subject(s)
Germ Layers/embryology , Germ Layers/metabolism , Ubiquitin-Protein Ligases/metabolism , X Chromosome Inactivation/genetics , Animals , Down-Regulation , Embryo Implantation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Histones/chemistry , Histones/metabolism , In Situ Hybridization, Fluorescence , Lysine/metabolism , Methylation , Mice , Mice, Knockout , RNA, Long Noncoding/genetics , Ubiquitin-Protein Ligases/genetics
16.
Biol Reprod ; 100(2): 440-454, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30239614

ABSTRACT

Mammalian sperm undergo a series of biochemical and physiological changes collectively known as capacitation in order to acquire the ability to fertilize. Although the increase in phosphorylation associated with mouse sperm capacitation is well established, the identity of the proteins involved in this signaling cascade remains largely unknown. Tandem mass spectrometry (MS/MS) has been used to identify the exact sites of phosphorylation and to compare the relative extent of phosphorylation at these sites. In the present work, we find that a novel site of phosphorylation on a peptide derived from the radial spoke protein Rsph6a is more phosphorylated in capacitated mouse sperm. The Rsph6a gene has six exons, five of which are conserved during evolution in flagellated cells. The exon containing the capacitation-induced phosphorylation site was found exclusively in eutherian mammals. Transcript analyses revealed at least two different testis-specific splicing variants for Rsph6a.Rsph6a mRNA expression was restricted to spermatocytes. Using antibodies generated against the Rsph6a N-terminal domain, western blotting and immunofluorescence analyses indicated that the protein remains in mature sperm and localizes to the sperm flagellum. Consistent with its role in the axoneme, solubility analyses revealed that Rsph6 is attached to cytoskeletal structures. Based on previous studies in Chlamydomonas reinhardtii, we predict that Rsph6 participates in the interaction between the central pair of microtubules and the surrounding pairs. The findings that Rsph6a is more phosphorylated during capacitation and is predicted to function in axonemal localization make Rsph6a a candidate protein mediating signaling processes in the sperm flagellum.


Subject(s)
Cytoskeletal Proteins/metabolism , Sperm Capacitation/physiology , Testis/metabolism , Animals , Antibodies , Cloning, Molecular , Cytoskeletal Proteins/genetics , Gene Expression Regulation/physiology , Male , Mice , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins
17.
Hepatology ; 68(1): 274-288, 2018 07.
Article in English | MEDLINE | ID: mdl-29315687

ABSTRACT

During development, the endoderm initiates organ-restricted gene expression patterns in a spatiotemporally controlled manner. This process, termed induction, requires signals from adjacent mesodermal derivatives. Fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) emanating from the cardiac mesoderm and the septum transversum mesenchyme (STM), respectively, are believed to be simultaneously and uniformly required to directly induce hepatic gene expression from the murine endoderm. Using small molecule inhibitors of BMP signals during liver bud induction in the developing mouse embryo, we found that BMP signaling was not uniformly required to induce hepatic gene expression. Although BMP inhibition caused an overall reduction in the number of induced hepatoblasts, the STM-bounded posterior liver bud demonstrated the most severe loss of the essential hepatic transcription factor, hepatocyte nuclear factor 4-α (HNF4α), whereas the sinus venosus-lined anterior liver bud was less affected. We found that the posterior liver bud progenitors were anteriorly displaced and aberrantly activated pancreatobiliary markers, including sex-determining region Y-box 9 (SOX9). Additionally, we found that ectopically expressed SOX9 inhibited HNF4α and that BMP was indirectly required for hepatoblast induction. Finally, because previous studies have demonstrated that FGF signals are essential for anterior but not posterior liver bud induction, we examined synchronous BMP and FGF inhibition and found this led to a nearly complete loss of hepatoblasts. CONCLUSION: BMP signaling is required to maintain the hepato-pancreatobiliary boundary, at least in part, by indirectly repressing SOX9 in the hepatic endoderm. BMP and FGF signals are each required for the induction of spatially complementary subsets of hepatoblasts. These results underscore the importance of studying early inductive processes in the whole embryo. (Hepatology 2018;68:274-288).


Subject(s)
Bone Morphogenetic Proteins/physiology , Embryonic Induction , Liver/embryology , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Cell Proliferation , Fibroblast Growth Factors/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Mice , SOX9 Transcription Factor/metabolism
18.
Reproduction ; 157(3): 215-222, 2019 03.
Article in English | MEDLINE | ID: mdl-30571656

ABSTRACT

Mediator is an evolutionarily conserved multi-subunit complex, bridging transcriptional activators and repressors to the general RNA polymerase II (Pol II) initiation machinery. Though the Mediator complex is crucial for the transcription of almost all Pol II promoters in eukaryotic organisms, the phenotypes of individual Mediator subunit mutants are each distinct. Here, we report for the first time, the essential role of subunit MED20 in early mammalian embryo development. Although Med20 mutant mouse embryos exhibit normal morphology at E3.5 blastocyst stage, they cannot be recovered at early post-gastrulation stages. Outgrowth assays show that mutant blastocysts cannot hatch from the zona pellucida, indicating impaired blastocyst function. Assessments of cell death and cell lineage specification reveal that apoptosis, inner cell mass, trophectoderm and primitive endoderm markers are normal in mutant blastocysts. However, the epiblast marker NANOG is ectopically expressed in the trophectoderm of Med20 mutants, indicative of defects in trophoblast specification. These results suggest that MED20 specifically, and the Mediator complex in general, are essential for the earliest steps of mammalian development and cell lineage specification.


Subject(s)
Blastocyst/cytology , Embryo, Mammalian/cytology , Embryonic Development , Gene Expression Regulation, Developmental , Mediator Complex/physiology , Nanog Homeobox Protein/genetics , Animals , Blastocyst/metabolism , Cell Lineage , Embryo, Mammalian/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanog Homeobox Protein/metabolism
19.
Biomacromolecules ; 20(1): 435-442, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30525500

ABSTRACT

RNA interference (RNAi) requires the intracellular delivery of RNA molecules to initiate the neutralization of targeted mRNA molecules, inhibiting the expression or translation of the targeted gene. Current polymers and lipids that are used to deliver RNA molecules are generally required to be positively charged, to achieve complexation with RNA and the cellular internalization. However, positive surface charge has been implicated as the reason for toxicity in many of these systems. Herein, we report a novel strategy to generate noncationic RNA-polymer complexes for RNA delivery with low cytotoxicity. We use an in situ electrostatic complexation using a methylated pyridinium group, which is simultaneously removed during the RNA binding step. The resultant complexes demonstrate successful knockdown in preimplantation mammalian embryos, thus providing a new approach for nucleic acid delivery.


Subject(s)
Gene Transfer Techniques , Nanoconjugates/chemistry , Polyelectrolytes/chemistry , RNA/chemistry , Animals , Cross-Linking Reagents/chemistry , Female , HeLa Cells , Humans , Mice , Nanoconjugates/adverse effects , Static Electricity
20.
Development ; 142(1): 31-40, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25516968

ABSTRACT

The maintenance and control of pluripotency is of great interest in stem cell biology. The dual specificity T-box/basic-helix-loop-helix-zipper transcription factor Mga is expressed in the pluripotent cells of the inner cell mass (ICM) and epiblast of the peri-implantation mouse embryo, but its function has not been investigated previously. Here, we use a loss-of-function allele and RNA knockdown to demonstrate that Mga depletion leads to the death of proliferating pluripotent ICM cells in vivo and in vitro, and the death of embryonic stem cells (ESCs) in vitro. Additionally, quiescent pluripotent cells lacking Mga are lost during embryonic diapause. Expression of Odc1, the rate-limiting enzyme in the conversion of ornithine into putrescine in the synthesis of polyamines, is reduced in Mga mutant cells, and the survival of mutant ICM cells as well as ESCs is rescued in culture by the addition of exogenous putrescine. These results suggest a mechanism whereby Mga influences pluripotent cell survival through regulation of the polyamine pool in pluripotent cells of the embryo, whether they are in a proliferative or quiescent state.


Subject(s)
Embryo Implantation , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Alleles , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors , Blastocyst Inner Cell Mass/cytology , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Crosses, Genetic , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Gene Knockdown Techniques , Gene Targeting , Genotype , Germ Layers/cytology , Germ Layers/metabolism , Male , Mice , Mutagenesis/genetics , Mutation/genetics , Ornithine Decarboxylase/metabolism , Pluripotent Stem Cells/metabolism , Polyamines/metabolism , Transcription Factors/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL