Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(10): 2092-2110.e23, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172563

ABSTRACT

The third and fourth weeks of gestation in primates are marked by several developmental milestones, including gastrulation and the formation of organ primordia. However, our understanding of this period is limited due to restricted access to in vivo embryos. To address this gap, we developed an embedded 3D culture system that allows for the extended ex utero culture of cynomolgus monkey embryos for up to 25 days post-fertilization. Morphological, histological, and single-cell RNA-sequencing analyses demonstrate that ex utero cultured monkey embryos largely recapitulated key events of in vivo development. With this platform, we were able to delineate lineage trajectories and genetic programs involved in neural induction, lateral plate mesoderm differentiation, yolk sac hematopoiesis, primitive gut, and primordial germ-cell-like cell development in monkeys. Our embedded 3D culture system provides a robust and reproducible platform for growing monkey embryos from blastocysts to early organogenesis and studying primate embryogenesis ex utero.


Subject(s)
Embryo, Mammalian , Embryonic Development , Animals , Macaca fascicularis , Blastocyst , Organogenesis , Primates
2.
Cell ; 186(26): 5859-5875.e24, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38052213

ABSTRACT

Embryogenesis necessitates harmonious coordination between embryonic and extraembryonic tissues. Although stem cells of both embryonic and extraembryonic origins have been generated, they are grown in different culture conditions. In this study, utilizing a unified culture condition that activates the FGF, TGF-ß, and WNT pathways, we have successfully derived embryonic stem cells (FTW-ESCs), extraembryonic endoderm stem cells (FTW-XENs), and trophoblast stem cells (FTW-TSCs) from the three foundational tissues of mouse and cynomolgus monkey (Macaca fascicularis) blastocysts. This approach facilitates the co-culture of embryonic and extraembryonic stem cells, revealing a growth inhibition effect exerted by extraembryonic endoderm cells on pluripotent cells, partially through extracellular matrix signaling. Additionally, our cross-species analysis identified both shared and unique transcription factors and pathways regulating FTW-XENs. The embryonic and extraembryonic stem cell co-culture strategy offers promising avenues for developing more faithful embryo models and devising more developmentally pertinent differentiation protocols.


Subject(s)
Embryo, Mammalian , Embryonic Stem Cells , Animals , Coculture Techniques , Macaca fascicularis , Embryonic Stem Cells/metabolism , Cell Differentiation , Endoderm/metabolism , Cell Lineage
3.
Cell ; 186(2): 287-304.e26, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36610399

ABSTRACT

Whether and how certain transposable elements with viral origins, such as endogenous retroviruses (ERVs) dormant in our genomes, can become awakened and contribute to the aging process is largely unknown. In human senescent cells, we found that HERVK (HML-2), the most recently integrated human ERVs, are unlocked to transcribe viral genes and produce retrovirus-like particles (RVLPs). These HERVK RVLPs constitute a transmissible message to elicit senescence phenotypes in young cells, which can be blocked by neutralizing antibodies. The activation of ERVs was also observed in organs of aged primates and mice as well as in human tissues and serum from the elderly. Their repression alleviates cellular senescence and tissue degeneration and, to some extent, organismal aging. These findings indicate that the resurrection of ERVs is a hallmark and driving force of cellular senescence and tissue aging.


Subject(s)
Aging , Endogenous Retroviruses , Aged , Animals , Humans , Mice , Aging/genetics , Aging/pathology , Cellular Senescence , Endogenous Retroviruses/genetics , Primates
4.
Cell ; 184(8): 2020-2032.e14, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33861963

ABSTRACT

Interspecies chimera formation with human pluripotent stem cells (hPSCs) represents a necessary alternative to evaluate hPSC pluripotency in vivo and might constitute a promising strategy for various regenerative medicine applications, including the generation of organs and tissues for transplantation. Studies using mouse and pig embryos suggest that hPSCs do not robustly contribute to chimera formation in species evolutionarily distant to humans. We studied the chimeric competency of human extended pluripotent stem cells (hEPSCs) in cynomolgus monkey (Macaca fascicularis) embryos cultured ex vivo. We demonstrate that hEPSCs survived, proliferated, and generated several peri- and early post-implantation cell lineages inside monkey embryos. We also uncovered signaling events underlying interspecific crosstalk that may help shape the unique developmental trajectories of human and monkey cells within chimeric embryos. These results may help to better understand early human development and primate evolution and develop strategies to improve human chimerism in evolutionarily distant species.


Subject(s)
Chimerism , Embryo, Mammalian/cytology , Pluripotent Stem Cells/cytology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryo, Mammalian/metabolism , Female , Humans , Macaca fascicularis , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , RNA-Seq , Single-Cell Analysis , Transcriptome
5.
Cell ; 169(5): 945-955.e10, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525759

ABSTRACT

Gene-editing technologies have made it feasible to create nonhuman primate models for human genetic disorders. Here, we report detailed genotypes and phenotypes of TALEN-edited MECP2 mutant cynomolgus monkeys serving as a model for a neurodevelopmental disorder, Rett syndrome (RTT), which is caused by loss-of-function mutations in the human MECP2 gene. Male mutant monkeys were embryonic lethal, reiterating that RTT is a disease of females. Through a battery of behavioral analyses, including primate-unique eye-tracking tests, in combination with brain imaging via MRI, we found a series of physiological, behavioral, and structural abnormalities resembling clinical manifestations of RTT. Moreover, blood transcriptome profiling revealed that mutant monkeys resembled RTT patients in immune gene dysregulation. Taken together, the stark similarity in phenotype and/or endophenotype between monkeys and patients suggested that gene-edited RTT founder monkeys would be of value for disease mechanistic studies as well as development of potential therapeutic interventions for RTT.


Subject(s)
Methyl-CpG-Binding Protein 2/genetics , Rett Syndrome/genetics , Animals , Brain/physiology , Chromosomes, Human, X , Circadian Rhythm , Disease Models, Animal , Electrocardiography , Female , Gene Editing , Humans , Macaca fascicularis , Magnetic Resonance Imaging , Male , Mutation , Pain , Rett Syndrome/physiopathology , Sleep , Transcription Activator-Like Effector Nucleases/metabolism , Transcriptome
7.
Cell ; 156(4): 836-43, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24486104

ABSTRACT

Monkeys serve as important model species for studying human diseases and developing therapeutic strategies, yet the application of monkeys in biomedical researches has been significantly hindered by the difficulties in producing animals genetically modified at the desired target sites. Here, we first applied the CRISPR/Cas9 system, a versatile tool for editing the genes of different organisms, to target monkey genomes. By coinjection of Cas9 mRNA and sgRNAs into one-cell-stage embryos, we successfully achieve precise gene targeting in cynomolgus monkeys. We also show that this system enables simultaneous disruption of two target genes (Ppar-γ and Rag1) in one step, and no off-target mutagenesis was detected by comprehensive analysis. Thus, coinjection of one-cell-stage embryos with Cas9 mRNA and sgRNAs is an efficient and reliable approach for gene-modified cynomolgus monkey generation.


Subject(s)
Gene Targeting/methods , Macaca fascicularis/genetics , Animals , Base Sequence , Cell Line , Embryo, Mammalian/metabolism , Female , Humans , Molecular Sequence Data , Mosaicism , Sequence Alignment
8.
Nature ; 577(7791): 537-542, 2020 01.
Article in English | MEDLINE | ID: mdl-31830756

ABSTRACT

Our understanding of how human embryos develop before gastrulation, including spatial self-organization and cell type ontogeny, remains limited by available two-dimensional technological platforms1,2 that do not recapitulate the in vivo conditions3-5. Here we report a three-dimensional (3D) blastocyst-culture system that enables human blastocyst development up to the primitive streak anlage stage. These 3D embryos mimic developmental landmarks and 3D architectures in vivo, including the embryonic disc, amnion, basement membrane, primary and primate unique secondary yolk sac, formation of anterior-posterior polarity and primitive streak anlage. Using single-cell transcriptome profiling, we delineate ontology and regulatory networks that underlie the segregation of epiblast, primitive endoderm and trophoblast. Compared with epiblasts, the amniotic epithelium shows unique and characteristic phenotypes. After implantation, specific pathways and transcription factors trigger the differentiation of cytotrophoblasts, extravillous cytotrophoblasts and syncytiotrophoblasts. Epiblasts undergo a transition to pluripotency upon implantation, and the transcriptome of these cells is maintained until the generation of the primitive streak anlage. These developmental processes are driven by different pluripotency factors. Together, findings from our 3D-culture approach help to determine the molecular and morphogenetic developmental landscape that occurs during human embryogenesis.


Subject(s)
Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development , Primitive Streak/cytology , Primitive Streak/embryology , Amnion/cytology , Amnion/embryology , Blastocyst/cytology , Cell Differentiation , Cell Lineage , Cell Polarity , Collagen , Drug Combinations , Epithelium/embryology , Gastrulation , Germ Layers/cytology , Germ Layers/embryology , Humans , Laminin , Proteoglycans , RNA-Seq , Single-Cell Analysis , Transcription Factors/metabolism , Transcriptome , Trophoblasts/cytology , Yolk Sac/cytology , Yolk Sac/embryology
9.
Neurobiol Dis ; 184: 106197, 2023 08.
Article in English | MEDLINE | ID: mdl-37328037

ABSTRACT

Poly(PR) is a dipeptide repeat protein comprising proline and arginine residues. It is one of the translational product of expanded G4C2 repeats in the C9orf72 gene, and its accumulation is contributing to the neuropathogenesis of C9orf72-associated amyotrophic lateral sclerosis and/or frontotemporal dementia (C9-ALS/FTD). In this study, we demonstrate that poly(PR) protein alone is sufficient to induce neurodegeneration related to ALS/FTD in cynomolgus monkeys. By delivering poly(PR) via AAV, we observed that the PR proteins were located within the nucleus of infected cells. The expression of (PR)50 protein, consisting of 50 PR repeats, led to increased loss of cortical neurons, cytoplasmic lipofuscin, and gliosis in the brain, as well as demyelination and loss of ChAT positive neurons in the spinal cord of monkeys. While, these pathologies were not observed in monkeys expressing (PR)5, a protein comprising only 5 PR repeats. Furthermore, the (PR)50-expressing monkeys exhibited progressive motor deficits, cognitive impairment, muscle atrophy, and abnormal electromyography (EMG) potentials, which closely resemble clinical symptoms seen in C9-ALS/FTD patients. By longitudinally tracking these monkeys, we found that changes in cystatin C and chitinase-1 (CHIT1) levels in the cerebrospinal fluid (CSF) corresponded to the phenotypic progression of (PR)50-induced disease. Proteomic analysis revealed that the major clusters of dysregulated proteins were nuclear-localized, and downregulation of the MECP2 protein was implicated in the toxic process of poly(PR). This research indicates that poly(PR) expression alone induces neurodegeneration and core phenotypes associated with C9-ALS/FTD in monkeys, which may provide insights into the mechanisms of disease pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Animals , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Amyotrophic Lateral Sclerosis/metabolism , Macaca fascicularis/genetics , Macaca fascicularis/metabolism , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Proteomics , Proteins/genetics , DNA Repeat Expansion , Dipeptides/genetics
10.
Neuroimage ; 247: 118799, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34896583

ABSTRACT

Longitudinal brain imaging atlases with densely sampled time-points and ancillary anatomical information are of fundamental importance in studying early developmental characteristics of human and non-human primate brains during infancy, which feature extremely dynamic imaging appearance, brain shape and size. However, for non-human primates, which are highly valuable animal models for understanding human brains, the existing brain atlases are mainly developed based on adults or adolescents, denoting a notable lack of temporally densely-sampled atlases covering the dynamic early brain development. To fill this critical gap, in this paper, we construct a comprehensive set of longitudinal brain atlases and associated tissue probability maps (gray matter, white matter, and cerebrospinal fluid) with totally 12 time-points from birth to 4 years of age (i.e., 1, 2, 3, 4, 5, 6, 9, 12, 18, 24, 36, and 48 months of age) based on 175 longitudinal structural MRI scans from 39 typically-developing cynomolgus macaques, by leveraging state-of-the-art computational techniques tailored for early developing brains. Furthermore, to facilitate region-based analysis using our atlases, we also provide two popular hierarchy parcellations, i.e., cortical hierarchy maps (6 levels) and subcortical hierarchy maps (6 levels), on our longitudinal macaque brain atlases. These early developing atlases, which have the densest time-points during infancy (to the best of our knowledge), will greatly facilitate the studies of macaque brain development.


Subject(s)
Brain/growth & development , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Animals , Gray Matter/growth & development , Image Processing, Computer-Assisted , Macaca fascicularis , White Matter/growth & development
11.
Biol Reprod ; 107(1): 237-249, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35766401

ABSTRACT

The induction of primordial germ-like cells (PGCLCs) from pluripotent stem cells (PSCs) provides a powerful system to study the cellular and molecular mechanisms underlying germline specification, which are difficult to study in vivo. The studies reveal the existence of a species-specific mechanism underlying PGCLCs between humans and mice, highlighting the necessity to study regulatory networks in more species, especially in primates. Harnessing the power of single-cell RNA sequencing (scRNA-seq) analysis, the detailed trajectory of human PGCLCs specification in vitro has been achieved. However, the study of nonhuman primates is still needed. Here, we applied an embryoid body (EB) differentiation system to induce PGCLCs specification from cynomolgus monkey male and female PSCs, and then performed high throughput scRNA-seq analysis of approximately 40 000 PSCs and cells within EBs. We found that EBs provided a niche for PGCLCs differentiation by secreting growth factors critical for PGCLC specification, such as bone morphogenetic protein 2 (BMP2), BMP4, and Wnt Family Member 3. Moreover, the developmental trajectory of PGCLCs was reconstituted, and gene expression dynamics were revealed. Our study outlines the roadmap of PGCLC specification from PSCs and provides insights that will improve the differentiation efficiency of PGCLCs from PSCs.


Subject(s)
Pluripotent Stem Cells , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Female , Germ Cells/metabolism , Humans , Macaca fascicularis/genetics , Male , Mice , RNA/metabolism
12.
Development ; 145(11)2018 06 11.
Article in English | MEDLINE | ID: mdl-29784672

ABSTRACT

Haploid embryonic stem cells (haESCs) have been extensively applied in forward and reverse genetic screening. However, a mammalian haploid somatic cell line is difficult to achieve because of spontaneous diploidization in differentiation. As a non-human primate species, monkeys are widely used in basic and pre-clinical research in which haploid cells are restricted to ESCs. Here, we report that rhesus monkey haESCs in an optimized culture medium show naïve-state pluripotency and stable haploidy. This model facilitated the derivation of haploid neural progenitor cells (haNPCs), which maintained haploidy and differentiation potential into neurons and glia for a long period in vitro High-throughput trapping mutations can be efficiently introduced into haNPCs via piggyBac transposons. This system proves useful when identifying gene targets of neural toxicants via a proof-of-concept experiment. Using CRISPR/Cas9 editing, we confirmed that B4GALT6, from the candidate gene list, is a resistance gene of A803467 (a tetrodotoxin-like toxicant). This model is the first non-human primate haploid somatic cell line with proliferative ability, multipotency and an intact genome, thus providing a cellular resource for recessive genetic and potential drug screening.


Subject(s)
Drug Evaluation, Preclinical/methods , Embryonic Stem Cells/cytology , Galactosyltransferases/genetics , Gene Editing/methods , Genetic Testing/veterinary , Macaca mulatta/embryology , Neural Stem Cells/cytology , Aniline Compounds/pharmacology , Animals , CRISPR-Cas Systems , DNA Transposable Elements/genetics , Furans/pharmacology , Genetic Testing/methods , Haploidy , Poisons/pharmacology
14.
J Biol Chem ; 293(30): 11654-11658, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29941452

ABSTRACT

CRISPR/Cas9 is now widely used in biomedical research and has great potential for clinical applications. However, the safety and efficacy of this gene-editing technique are significant issues. Recent reports on mouse models and human cells have raised concerns that off-target mutations could hamper applying the CRISPR technology in patients. The high similarities of nonhuman primates to humans in genome content and organization, genetic diversity, physiology, and cognitive abilities have made these animals ideal experimental models for understanding human diseases and developing therapeutics. Off-target mutations of CRISPR/Cas9 have been analyzed in previous studies of nonhuman primates, but no report has investigated genome-wide off-target effects in living monkeys. Here, we used rhesus monkeys in which a genetic disorder mimicking Duchenne muscular dystrophy had previously been produced with CRISPR/Cas9. Using whole-genome sequencing to comprehensively assess on- and off-target mutations in these animals, we found that CRISPR/Cas9-based gene editing is active on the expected genomic sites without producing off-target modifications in other functional regions of the genome. These findings suggest that the CRISPR/Cas9 technique could be relatively safe and effective in modeling genetic disease in nonhuman primates and in future therapeutic research of human diseases.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Muscular Dystrophy, Duchenne/genetics , Mutation , Whole Genome Sequencing , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Mutational Analysis/methods , Disease Models, Animal , Gene Editing/methods , Gene Knockout Techniques/methods , Gene Library , Macaca mulatta , Whole Genome Sequencing/methods
15.
BMC Biotechnol ; 19(1): 7, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646876

ABSTRACT

BACKGROUND: Non-human primate (NHP) models can closely mimic human physiological functions and are therefore highly valuable in biomedical research. Genome editing is now developing rapidly due to the precision and efficiency offered by engineered site-specific endonuclease-based systems, such as transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 nuclease (Cas9) system. It has been demonstrated that these programmable nucleases can introduce genetic changes in embryos from many species including NHPs. In 2014, we reported the first genetic editing of macaques using TALENs and CRISPR/Cas9. Subsequently, we characterized the phenotype of a methyl CpG binding protein 2 (MECP2)-mutant cynomolgus monkey model of Rett syndrome generated using the TALEN approach. These efforts not only accelerated the advance of modeling genetic diseases in NHPs, but also encouraged us to develop specific gene knock-in monkeys. In this study, we assess the possibility of homologous recombination (HR)-mediated gene replacement using TALENs in monkeys, and generate preimplantation embryos carrying an EmGFP fluorescent reporter constructed in the OCT4 gene. RESULT: We assembled a pair of TALENs specific to the first exon of the OCT4 gene and constructed a donor vector consisting of the homology arms cloned from the monkey genome DNA, flanking an EmGFP cassette. Next, we co-injected the TALENs-coding plasmid and donor plasmid into the cytoplasm of 122 zygotes 6-8 h after fertilization. Sequencing and immunofluorescence revealed that the OCT4-EmGFP knock-in allele had been successfully generated by TALENs-mediated HR at an efficiency of 11.3% (7 out of 62) or 11.1% (1 out of 9), respectively, in monkey embryos. CONCLUSION: We have successfully, for the first time, obtained OCT4-EmGFP knock-in monkey embryos via HR mediated by TALENs. Our results suggest that gene targeting through TALEN-assisted HR is a useful approach to introduce precise genetic modification in NHPs.


Subject(s)
Gene Editing/methods , Gene Knock-In Techniques/methods , Homologous Recombination , Macaca fascicularis/embryology , Macaca fascicularis/genetics , Transcription Activator-Like Effector Nucleases/genetics , Animals , Animals, Genetically Modified , Female , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism
16.
Clin Proteomics ; 16: 24, 2019.
Article in English | MEDLINE | ID: mdl-31244561

ABSTRACT

BACKGROUND: Although sperm cryopreservation has been widely used in human reproductive medicine as an integral infertility management in infertility clinics and for banking sperm in sperm banks, the freezing/thawing protocols are not optimal. The freezing and thawing processes result in changes at both structural and molecular levels, some even detrimental, in human sperm when compared with fresh sperm. The change of sperm proteins after cryopreservation may play negative roles for fertilization and early embryo development. Conventionally, cryostraws (CS) and cryovials (CV) are the most widely used cryopreservation carriers (CPCs) for human sperm cryopreservation accompanied with the use of egg yolk free commercial media. However, the influence of cryopreservation on the proteomic profile of human sperm preserved with the two CPCs is unknown. Therefore the purpose of the present study was to compare the frozen-thawed motility, investigate the proteomic profile of human sperm cryopreserved with the two types of CPCs, and identify the susceptible proteins that play key roles for sperm function and fertility. METHODS: The present study compared the cryosurvival of human sperm frozen with the two different CPCs and identified the sperm proteomic changes by using the isobaric tags for relative and absolute quantification labeling technique coupled with 2D LC-MS/MS analysis after freezing and thawing. RESULTS: Our results indicated that sperm cryopreserved with CV showed higher values for percentage of motile sperm and forward activity rate than those with CS. Compared to fresh sperm, 434 and 432 proteins were differentially identified in human sperm cryopreserved with CS and CV, respectively. CONCLUSION: The proteomic profiles of human sperm are greatly affected by cryopreservation with either type of CPC. GO analysis revealed that most of the differentially identified sperm proteins enriched in the extracellular membrane-bounded organelles, cytoplasm and cytosol. In addition, 106 susceptible proteins having known identities related to sperm functions were identified. In general, cryovial seems to be the preferred CPC for human sperm cryopreservation based on the post-thaw motility parameters and the effect on sperm proteomic profiles. These results are beneficial for the insight into the understanding of the cryoinjury mechanism of sperm and the development of human sperm cryopreservation strategies.

17.
Hum Mol Genet ; 24(25): 7255-64, 2015 Dec 20.
Article in English | MEDLINE | ID: mdl-26464492

ABSTRACT

Mutations in the DAX1 locus cause X-linked adrenal hypoplasia congenita (AHC) and hypogonadotropic hypogonadism (HH), which manifest with primary adrenal insufficiency and incomplete or absent sexual maturation, respectively. The associated defects in spermatogenesis can range from spermatogenic arrest to Sertoli cell only syndrome. Conclusions from Dax1 knockout mouse models provide only limited insight into AHC/HH disease mechanisms, because mouse models exhibit more extensive abnormalities in testicular development, including disorganized and incompletely formed testis cords with decreased number of peritubular myoid cells and male-to-female sex reversal. We previously reported successful clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated genome targeting in cynomolgus monkeys. Here, we describe a male fetal monkey in which targeted genome editing using CRISPR/Cas9 produced Dax1-null mutations in most somatic tissues and in the gonads. This DAX1-deficient monkey displayed defects in adrenal gland development and abnormal testis architecture with small cords, expanded blood vessels and extensive fibrosis. Sertoli cell formation was not affected. This phenotype strongly resembles findings in human patients with AHC-HH caused by mutations in DAX1. We further detected upregulation of Wnt/ß-catenin-VEGF signaling in the fetal Dax1-deficient testis, suggesting abnormal activation of signaling pathways in the absence of DAX1 as one mechanism of AHC-HH. Our study reveals novel insight into the role of DAX1 in HH and provides proof-of-principle for the generation of monkey models of human disease via CRISPR/Cas9-mediated gene targeting.


Subject(s)
CRISPR-Associated Proteins/metabolism , Testis/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , CRISPR-Associated Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Haplorhini , Humans , Hypogonadism/genetics , Hypogonadism/metabolism , Male , Sertoli Cells/metabolism , Transcription Factors/genetics
18.
Hum Mol Genet ; 24(8): 2308-17, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25552648

ABSTRACT

Parkinson's disease (PD) is an age-dependent neurodegenerative disease that can be caused by genetic mutations in α-synuclein (α-syn) or duplication of wild-type α-syn; PD is characterized by the deposition of α-syn aggregates, indicating a gain of toxicity from accumulation of α-syn. Although the major neuropathologic feature of PD is the degeneration of dopaminergic (DA) neurons in the substantia nigra, non-motor symptoms including anxiety, cognitive defect and sleep disorder precede the onset of motor impairment, and many clinical symptoms of PD are not caused by degeneration of DA neurons. Non-human primate models of PD are important for revealing the early pathology in PD and identifying effective treatments. We established transgenic PD rhesus monkeys that express mutant α-syn (A53T). Six transgenic A53T monkeys were produced via lentiviral vector expressing A53T in fertilized monkey eggs and subsequent embryo transfer to surrogates. Transgenic A53T is expressed in the monkey brain and causes age-dependent non-motor symptoms, including cognitive defects and anxiety phenotype, without detectable sleeping disorders. The transgenic α-syn monkeys demonstrate the specific early symptoms caused by mutant α-syn and provide insight into treatment of early PD.


Subject(s)
Disease Models, Animal , Macaca mulatta , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Brain/metabolism , Brain/pathology , Dopaminergic Neurons/metabolism , Female , Humans , Macaca mulatta/genetics , Macaca mulatta/metabolism , Male , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity
19.
Hum Mol Genet ; 24(13): 3764-74, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25859012

ABSTRACT

CRISPR/Cas9 has been used to genetically modify genomes in a variety of species, including non-human primates. Unfortunately, this new technology does cause mosaic mutations, and we do not yet know whether such mutations can functionally disrupt the targeted gene or cause the pathology seen in human disease. Addressing these issues is necessary if we are to generate large animal models of human diseases using CRISPR/Cas9. Here we used CRISPR/Cas9 to target the monkey dystrophin gene to create mutations that lead to Duchenne muscular dystrophy (DMD), a recessive X-linked form of muscular dystrophy. Examination of the relative targeting rate revealed that Crispr/Cas9 targeting could lead to mosaic mutations in up to 87% of the dystrophin alleles in monkey muscle. Moreover, CRISPR/Cas9 induced mutations in both male and female monkeys, with the markedly depleted dystrophin and muscle degeneration seen in early DMD. Our findings indicate that CRISPR/Cas9 can efficiently generate monkey models of human diseases, regardless of inheritance patterns. The presence of degenerated muscle cells in newborn Cas9-targeted monkeys suggests that therapeutic interventions at the early disease stage may be effective at alleviating the myopathy.


Subject(s)
Disease Models, Animal , Dystrophin/genetics , Endonucleases/metabolism , Gene Targeting/methods , Macaca mulatta/genetics , Muscular Dystrophy, Duchenne/genetics , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Dystrophin/metabolism , Female , Humans , Macaca mulatta/metabolism , Male , Muscular Dystrophy, Duchenne/metabolism , Mutation
20.
Biol Reprod ; 97(5): 758-761, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29069285

ABSTRACT

Intracytoplasmic sperm injection (ICSI) and embryo transfer (ET) in nonhuman primates, e.g. rhesus and cynomolgus monkeys, has been widely used in researches of reproductive and developmental biology, and the success rate has been improved significantly. However, unwanted multiple pregnancy occurs frequently during the ICSI-ET in monkeys, most of which leads to miscarriages. To improve the birth rate of pregnancies and to safeguard health of host and baby monkeys, multifetal pregnancy reduction (MPR) is necessary. In this study, a total of 10 monkeys with multiple pregnancies received MPR through transabdominal ultrasound-guided potassium chloride injection into beating hearts of selective fetuses. To assess MPR efficiency, 31 monkeys with normal singleton pregnancies and 25 monkeys with twin pregnancies without MPR were used as controls. The aim of the reduction is to keep only one fetus, no matter twin or triplet pregnancy originally. Our results show that six cases of MPR were successful and all of them retained single fetus. Moreover, about 1 month (30.2 ± 1.2 days) of gestation is a better timing for MPR than later stage (50.7 ± 1.9 days). We also found that the remaining fetuses developed normally with full-term gestation and normal birth weight. In conclusion, transabdominal ultrasound-guided potassium chloride injection is a safe and effective MPR method for monkeys with multiple pregnancies.


Subject(s)
Haplorhini , Pregnancy Reduction, Multifetal/veterinary , Pregnancy, Animal , Pregnancy, Multiple , Abortion, Spontaneous , Animals , Embryo Transfer/veterinary , Female , Pregnancy , Pregnancy Outcome , Pregnancy Reduction, Multifetal/methods
SELECTION OF CITATIONS
SEARCH DETAIL