Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
EMBO J ; 41(18): e110815, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35912849

ABSTRACT

In vitro oogenesis is key to elucidating the mechanism of human female germ-cell development and its anomalies. Accordingly, pluripotent stem cells have been induced into primordial germ cell-like cells and into oogonia with epigenetic reprogramming, yet further reconstitutions remain a challenge. Here, we demonstrate ex vivo reconstitution of fetal oocyte development in both humans and cynomolgus monkeys (Macaca fascicularis). With an optimized culture of fetal ovary reaggregates over three months, human and monkey oogonia enter and complete the first meiotic prophase to differentiate into diplotene oocytes that form primordial follicles, the source for oogenesis in adults. The cytological and transcriptomic progressions of fetal oocyte development in vitro closely recapitulate those in vivo. A comparison of single-cell transcriptomes among humans, monkeys, and mice unravels primate-specific and conserved programs driving fetal oocyte development, the former including a distinct transcriptomic transformation upon oogonia-to-oocyte transition and the latter including two active X chromosomes with little X-chromosome upregulation. Our study provides a critical step forward for realizing human in vitro oogenesis and uncovers salient characteristics of fetal oocyte development in primates.


Subject(s)
Meiosis , Oogenesis , Animals , Female , Humans , Macaca fascicularis , Mice , Oocytes , Oogenesis/physiology , Ovary
2.
Biol Reprod ; 100(6): 1440-1452, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30869744

ABSTRACT

Nonhuman primates (NHPs) are considered to be the most valuable models for human transgenic (Tg) research into disease because human pathology is more closely recapitulated in NHPs than rodents. Previous studies have reported the generation of Tg NHPs that ubiquitously overexpress a transgene using various promoters, but it is not yet clear which promoter is most suitable for the generation of NHPs overexpressing a transgene ubiquitously and persistently in various tissues. To clarify this issue, we evaluated four putative ubiquitous promoters, cytomegalovirus (CMV) immediate-early enhancer and chicken beta-actin (CAG), elongation factor 1α (EF1α), ubiquitin C (UbC), and CMV, using an in vitro differentiation system of cynomolgus monkey embryonic stem cells (ESCs). While the EF1α promoter drove Tg expression more strongly than the other promoters in undifferentiated pluripotent ESCs, the CAG promoter was more effective in differentiated cells such as embryoid bodies and ESC-derived neurons. When the CAG and EF1α promoters were used to generate green fluorescent protein (GFP)-expressing Tg monkeys, the CAG promoter drove GFP expression in skin and hematopoietic tissues more strongly than in ΕF1α-GFP Tg monkeys. Notably, the EF1α promoter underwent more silencing in both ESCs and Tg monkeys. Thus, the CAG promoter appears to be the most suitable for ubiquitous and stable expression of transgenes in the differentiated tissues of Tg cynomolgus monkeys and appropriate for the establishment of human disease models.


Subject(s)
Animals, Genetically Modified , Genetic Vectors , Macaca fascicularis/genetics , Promoter Regions, Genetic , Transgenes , Actins/genetics , Animals , Antigens, Viral/genetics , Cells, Cultured , Chickens/genetics , Cloning, Organism/methods , Cloning, Organism/standards , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Enhancer Elements, Genetic/genetics , Female , Gene Transfer Techniques/standards , Genetic Vectors/genetics , Immediate-Early Proteins/genetics , Male , Mice , Peptide Elongation Factor 1/genetics
3.
J Reprod Dev ; 63(2): 167-174, 2017 Apr 21.
Article in English | MEDLINE | ID: mdl-28190810

ABSTRACT

The fertilized oocyte begins cleavage, leading to zygotic gene activation (ZGA), which re-activates the resting genome to acquire totipotency. In this process, genomic function is regulated by the dynamic structural conversion in the nucleus. Indeed, a considerable number of genes that are essential for embryonic development are located near the pericentromeric regions, wherein the heterochromatin is formed. These genes are repressed transcriptionally in somatic cells. Three-dimensional fluorescence in situ hybridization (3D-FISH) enables the visualization of the intranuclear spatial arrangement, such as gene loci, chromosomal domains, and chromosome territories (CTs). However, the 3D-FISH approach in mammalian embryos has been limited to certain repeated sequences because of its unfavorable properties. In this study, we developed an easy-to-use chamber device (EASI-FISH chamber) for 3D-FISH in early embryos, and visualized, for the first time, the spatial arrangements of pericentromeric regions, the ZGA-activated gene (Zscan4) loci, and CTs (chromosome 7), simultaneously during the early cleavage stage of mouse embryos by 3D-FISH. As a result, it was revealed that morphological changes of the pericentromeric regions and CTs, and relocation of the Zscan4 loci in CTs, occurred in the 1- to 4-cell stage embryos, which was different from those in somatic cells. This convenient and reproducible 3D-FISH technique for mammalian embryos represents a valuable tool that will provide insights into the nuclear dynamics of development.


Subject(s)
Cell Nucleus/genetics , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization, Fluorescence/methods , Animals , Female , Mice , Pregnancy
4.
Sci Rep ; 13(1): 15649, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37730951

ABSTRACT

Familial hypercholesterolemia (FH) is an inherited autosomal dominant disorder that is associated with a high plasma level of low-density lipoprotein (LDL) cholesterol, leading to an increased risk of cardiovascular diseases. To develop basic and translational research on FH, we here generated an FH model in a non-human primate (cynomolgus monkeys) by deleting the LDL receptor (LDLR) gene using the genome editing technique. Six LDLR knockout (KO) monkeys were produced, all of which were confirmed to have mutations in the LDLR gene by sequence analysis. The levels of plasma cholesterol and triglyceride were quite high in the monkeys, and were similar to those in FH patients with homozygous mutations in the LDLR gene. In addition, periocular xanthoma was observed only 1 year after birth. Lipoprotein profile analysis showed that the plasma very low-density lipoprotein and LDL were elevated, while the plasma high density lipoprotein was decreased in LDLR KO monkeys. The LDLR KO monkeys were also strongly resistant to medications for hypercholesterolemia. Taken together, we successfully generated a non-human primate model of hypercholesterolemia in which the phenotype is similar to that of homozygous FH patients.


Subject(s)
Craniocerebral Trauma , Hypercholesterolemia , Hyperlipoproteinemia Type II , Animals , Humans , Primates , Hyperlipoproteinemia Type II/genetics , Lipoproteins, LDL , Macaca fascicularis
5.
Sci Data ; 8(1): 159, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34183680

ABSTRACT

Cynomolgus macaque (Macaca fascicularis) and common marmoset (Callithrix jacchus) have been widely used in human biomedical research. Long-standing primate genome assemblies used the human genome as a reference for ordering and orienting the assembled fragments into chromosomes. Here we performed de novo genome assembly of these two species without any human genome-based bias observed in the genome assemblies released earlier. We assembled PacBio long reads, and the resultant contigs were scaffolded with Hi-C data, which were further refined based on Hi-C contact maps and alternate de novo assemblies. The assemblies achieved scaffold N50 lengths of 149 Mb and 137 Mb for cynomolgus macaque and common marmoset, respectively. The high fidelity of our assembly is also ascertained by BAC-end concordance in common marmoset. Our assembly of cynomolgus macaque outperformed all the available assemblies of this species in terms of contiguity. The chromosome-scale genome assemblies produced in this study are valuable resources for non-human primate models and provide an important baseline in human biomedical research.


Subject(s)
Callithrix/genetics , Contig Mapping , Macaca fascicularis/genetics , Animals , Chromosomes , Gene Order
6.
Life Sci Alliance ; 4(5)2021 05.
Article in English | MEDLINE | ID: mdl-33608411

ABSTRACT

The in vitro reconstitution of human germ-cell development provides a robust framework for clarifying key underlying mechanisms. Here, we explored transcription factors (TFs) that engender the germ-cell fate in their pluripotent precursors. Unexpectedly, SOX17, TFAP2C, and BLIMP1, which act under the BMP signaling and are indispensable for human primordial germ-cell-like cell (hPGCLC) specification, failed to induce hPGCLCs. In contrast, GATA3 or GATA2, immediate BMP effectors, combined with SOX17 and TFAP2C, generated hPGCLCs. GATA3/GATA2 knockouts dose-dependently impaired BMP-induced hPGCLC specification, whereas GATA3/GATA2 expression remained unaffected in SOX17, TFAP2C, or BLIMP1 knockouts. In cynomolgus monkeys, a key model for human development, GATA3, SOX17, and TFAP2C were co-expressed exclusively in early PGCs. Crucially, the TF-induced hPGCLCs acquired a hallmark of bona fide hPGCs to undergo epigenetic reprogramming and mature into oogonia/gonocytes in xenogeneic reconstituted ovaries. By uncovering a TF circuitry driving the germ line program, our study provides a paradigm for TF-based human gametogenesis.


Subject(s)
Germ Cells/metabolism , SOXF Transcription Factors/metabolism , Transcription Factor AP-2/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Female , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Germ Cells/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Macaca fascicularis , Mice , Mice, Inbred ICR , SOXF Transcription Factors/genetics , Signal Transduction/genetics , Transcription Factor AP-2/genetics , Transcription Factors/metabolism
7.
J Alzheimers Dis ; 75(1): 45-60, 2020.
Article in English | MEDLINE | ID: mdl-32250299

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia and understanding its pathogenesis should lead to improved therapeutic and diagnostic methods. Although several groups have developed transgenic mouse models overexpressing the human amyloid-ß precursor protein (APP) gene with AD mutations, with and without presenilin mutations, as well as APP gene knock-in mouse models, these animals display amyloid pathology but do not show neurofibrillary tangles or neuronal loss. This presumably is due to differences between the etiology of the aged-related human disease and the mouse models. Here we report the generation of two transgenic cynomolgus monkeys overexpressing the human gene for APP with Swedish, Artic, and Iberian mutations, and demonstrated expression of gene tagged green fluorescent protein marker in the placenta, amnion, hair follicles, and peripheral blood. We believe that these nonhuman primate models will be very useful to study the pathogenesis of dementia and AD. However, generated Tg monkeys still have some limitations. We employed the CAG promoter, which will promote gene expression in a non-tissue specific manner. Moreover, we used transgenic models but not knock-in models. Thus, the inserted transgene destroys endogenous gene(s) and may affect the phenotype(s). Nevertheless, it will be of great interest to determine whether these Tg monkeys will develop tauopathy and neurodegeneration similar to human AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Macaca fascicularis/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Promoter Regions, Genetic
8.
Stem Cell Res ; 37: 101439, 2019 05.
Article in English | MEDLINE | ID: mdl-31005787

ABSTRACT

Cynomolgus monkey ES (Cyn ES) cells can be generated in a similar manner as human ES cells. However, Cyn ES cells are difficult to maintain in an undifferentiated state by untrained researchers. For easier culture, we generated an OCT3/4-P2A tdTomato IRES ZeocinR Cyn ES cell line using CRISPR/Cas9 genome editing technology. The stop codon of the endogenous OCT3/4 locus was replaced with the P2A tdTomato IRES ZeocinR pA cassette by homologous recombination. This cell line enables us to isolate pluripotent stem cells and exclude differentiated cells by addition of zeocin, especially for culture without feeder cells.


Subject(s)
CRISPR-Cas Systems , Cell Differentiation , Cell Separation/methods , Embryonic Stem Cells/cytology , Gene Editing , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/genetics , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Feeder Cells , Homologous Recombination , Induced Pluripotent Stem Cells/metabolism , Macaca fascicularis , Octamer Transcription Factor-3/antagonists & inhibitors
9.
Nat Commun ; 10(1): 5517, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31822676

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) caused by PKD1 mutations is one of the most common hereditary disorders. However, the key pathological processes underlying cyst development and exacerbation in pre-symptomatic stages remain unknown, because rodent models do not recapitulate critical disease phenotypes, including disease onset in heterozygotes. Here, using CRISPR/Cas9, we generate ADPKD models with PKD1 mutations in cynomolgus monkeys. As in humans and mice, near-complete PKD1 depletion induces severe cyst formation mainly in collecting ducts. Importantly, unlike in mice, PKD1 heterozygote monkeys exhibit cyst formation perinatally in distal tubules, possibly reflecting the initial pathology in humans. Many monkeys in these models survive after cyst formation, and cysts progress with age. Furthermore, we succeed in generating selective heterozygous mutations using allele-specific targeting. We propose that our models elucidate the onset and progression of ADPKD, which will serve as a critical basis for establishing new therapeutic strategies, including drug treatments.


Subject(s)
Macaca fascicularis , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Alleles , Animals , Disease Models, Animal , Female , Heterozygote , Humans , Kidney/metabolism , Kidney/pathology , Macaca fascicularis/genetics , Macaca fascicularis/metabolism , Male , Mutation , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , TRPP Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL