Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 109(6): 812-820, 2023 12 11.
Article in English | MEDLINE | ID: mdl-37688580

ABSTRACT

Embryo morphokinetic analysis through time-lapse embryo imaging is envisioned as a method to improve selection of developmentally competent embryos. Morphokinetic analysis could be utilized to evaluate the effects of experimental manipulation on pre-implantation embryo development. The objectives of this study were to establish a normative morphokinetic database for in vitro fertilized rhesus macaque embryos and to assess the impact of atypical initial cleavage patterns on subsequent embryo development and formation of embryo outgrowths. The cleavage pattern and the timing of embryo developmental events were annotated retrospectively for unmanipulated in vitro fertilized rhesus macaque blastocysts produced over four breeding seasons. Approximately 50% of the blastocysts analyzed had an abnormal early cleavage event. The time to the initiation of embryo compaction and the time to completion of hatching was significantly delayed in blastocysts with an abnormal early cleavage event compared to blastocysts that had cleaved normally. Embryo hatching, attachment to an extracellular matrix, and growth during the implantation stage in vitro was not impacted by the initial cleavage pattern. These data establish normative morphokinetic parameters for in vitro fertilized rhesus macaque embryos and suggest that cleavage anomalies may not impact embryo implantation rates following embryo transfer.


Subject(s)
Embryonic Development , Fertilization in Vitro , Animals , Macaca mulatta , Retrospective Studies , Fertilization in Vitro/veterinary , Fertilization in Vitro/methods , Embryo, Mammalian , Embryo Implantation , Blastocyst , Time-Lapse Imaging/methods , Embryo Culture Techniques/veterinary , Embryo Culture Techniques/methods
2.
Retrovirology ; 19(1): 17, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35948929

ABSTRACT

Nonhuman primates (NHPs) are well-established basic and translational research models for human immunodeficiency virus (HIV) infections and pathophysiology, hematopoietic stem cell (HSC) transplantation, and assisted reproductive technologies. Recent advances in CRISPR/Cas9 gene editing technologies present opportunities to refine NHP HIV models for investigating genetic factors that affect HIV replication and designing cellular therapies that exploit genetic barriers to HIV infections, including engineering mutations into CCR5 and conferring resistance to HIV/simian immunodeficiency virus (SIV) infections. In this report, we provide an overview of recent advances and challenges in gene editing NHP embryos and discuss the value of genetically engineered animal models for developing novel stem cell-based therapies for curing HIV.


Subject(s)
Gene Editing , HIV Infections , Animals , CRISPR-Cas Systems , Humans , Primates , Stem Cells
3.
Sci Rep ; 12(1): 7348, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35513694

ABSTRACT

Zika virus (ZIKV) infection at the maternal-placental interface is associated with adverse pregnancy outcomes including fetal demise and pregnancy loss. To determine how infection impacts placental trophoblasts, we utilized rhesus macaque trophoblast stem cells (TSC) that can be differentiated into early gestation syncytiotrophoblasts (ST) and extravillous trophoblasts (EVT). TSCs and STs, but not EVTs, were highly permissive to productive infection with ZIKV strain DAK AR 41524. The impact of ZIKV on the cellular transcriptome showed that infection of TSCs and STs increased expression of immune related genes, including those involved in type I and type III interferon responses. ZIKV exposure altered extracellular vesicle (EV) mRNA, miRNA and protein cargo, including ZIKV proteins, regardless of productive infection. These findings suggest that early gestation macaque TSCs and STs are permissive to ZIKV infection, and that EV analysis may provide a foundation for identifying non-invasive biomarkers of placental infection in a highly translational model.


Subject(s)
Extracellular Vesicles , Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus , Animals , Extracellular Vesicles/genetics , Female , Gene Expression , Humans , Macaca mulatta , Placenta/metabolism , Pregnancy , Trophoblasts/metabolism , Zika Virus/physiology
4.
Front Genome Ed ; 4: 1031275, 2022.
Article in English | MEDLINE | ID: mdl-36714391

ABSTRACT

Introduction: Genome editing by CRISPR-Cas9 approaches offers promise for introducing or correcting disease-associated mutations for research and clinical applications. Nonhuman primates are physiologically closer to humans than other laboratory animal models, providing ideal candidates for introducing human disease-associated mutations to develop models of human disease. The incidence of large chromosomal anomalies in CRISPR-Cas9-edited human embryos and cells warrants comprehensive genotypic investigation of editing outcomes in primate embryos. Our objective was to evaluate on- and off-target editing outcomes in CCR5 CRISPR-Cas9-targeted Mauritian cynomolgus macaque embryos. Methods: DNA isolated from individual blastomeres of two embryos, along with paternal and maternal DNA, was subjected to whole genome sequencing (WGS) analysis. Results: Large deletions were identified in macaque blastomeres at the on-target site that were not previously detected using PCR-based methods. De novo mutations were also identified at predicted CRISPR-Cas9 off-target sites. Discussion: This is the first report of WGS analysis of CRISPR-Cas9-targeted nonhuman primate embryonic cells, in which a high editing efficiency was coupled with the incidence of editing errors in cells from two embryos. These data demonstrate that comprehensive sequencing-based methods are warranted for evaluating editing outcomes in primate embryos, as well as any resultant offspring to ensure that the observed phenotype is due to the targeted edit and not due to unidentified off-target mutations.

5.
Biol Reprod ; 104(1): 27-57, 2021 01 04.
Article in English | MEDLINE | ID: mdl-32856695

ABSTRACT

Monitoring the health of a pregnancy is of utmost importance to both the fetus and the mother. The diagnosis of pregnancy complications typically occurs after the manifestation of symptoms, and limited preventative measures or effective treatments are available. Traditionally, pregnancy health is evaluated by analyzing maternal serum hormone levels, genetic testing, ultrasonographic imaging, and monitoring maternal symptoms. However, researchers have reported a difference in extracellular vesicle (EV) quantity and cargo between healthy and at-risk pregnancies. Thus, placental EVs (PEVs) may help to understand normal and aberrant placental development, monitor pregnancy health in terms of developing placental pathologies, and assess the impact of environmental influences, such as infection, on pregnancy. The diagnostic potential of PEVs could allow for earlier detection of pregnancy complications via noninvasive sampling and frequent monitoring. Understanding how PEVs serve as a means of communication with maternal cells and recognizing their potential utility as a readout of placental health have sparked a growing interest in basic and translational research. However, to date, PEV research with animal models lags behind human studies. The strength of animal pregnancy models is that they can be used to assess placental pathologies in conjunction with isolation of PEVs from fluid samples at different time points throughout gestation. Assessing PEV cargo in animals within normal and complicated pregnancies will accelerate the translation of PEV analysis into the clinic for potential use in prognostics. We propose that appropriate animal models of human pregnancy complications must be established in the PEV field.


Subject(s)
Extracellular Vesicles/metabolism , Placenta Diseases/diagnosis , Placenta/metabolism , Exosomes/metabolism , Female , Humans , Placenta Diseases/metabolism , Pregnancy
6.
Sci Rep ; 10(1): 19159, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154556

ABSTRACT

Nonhuman primates are excellent models for studying human placentation as experimental manipulations in vitro can be translated to in vivo pregnancy. Our objective was to develop macaque trophoblast stem cells (TSCs) as an in vitro platform for future assessment of primate trophoblast development and function. Macaque TSC lines were generated by isolating first and second trimester placental villous cytotrophoblasts followed by culture in TSC medium to maintain cellular proliferation. TSCs grew as mononuclear colonies, whereas upon induction of syncytiotrophoblast (ST) differentiation multinuclear structures appeared, indicative of syncytium formation. Chorionic gonadotropin secretion was > 4000-fold higher in ST culture media compared to TSC media. The secretion of chorionic gonadotropin by TSC-derived ST reflects a reprogramming of macaque TSCs to an earlier pregnancy phenotype. Characteristic trophoblast hallmarks were defined in TSCs and ST including expression of C19MC miRNAs and the macaque placental nonclassical MHC class I molecule, Mamu-AG. Extravillous trophoblasts (EVTs) were derived that express macaque EVT markers Mamu-AG and CD56, and also secrete high levels of MMP2. Our analyses of macaque TSCs suggests that these cells represent a proliferative, self-renewing population capable of differentiating to STs and EVTs in vitro thereby establishing an experimental model of primate placentation.


Subject(s)
Cell Differentiation/physiology , Phenotype , Placenta/cytology , Stem Cells/cytology , Trophoblasts/cytology , Animals , Cell Proliferation/physiology , Female , Macaca , Placentation/physiology , Pregnancy
7.
Sci Rep ; 10(1): 18457, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33116147

ABSTRACT

The discovery that CCR5 serves as an R5-HIV-1 co-receptor, coupled with findings of protection from HIV infection in individuals lacking CCR5, led to the exploration of novel therapeutic strategies for HIV infection based on genome editing of CCR5. Advancing translation of CCR5-mutant-based cellular therapies for HIV requires development of novel physiologically relevant animal models. Mauritian cynomolgus macaques (MCMs), with high degree of MHC allele sharing, are valuable models for HIV-1 research and stem cell therapies. To facilitate the generation of a CCR5-mutant MHC-defined MCM model, we explored editing the CCR5 gene in MCM embryos via CRISPR-Cas9. We refined ovarian stimulation and in vitro fertilization (IVF) methods established for Chinese cynomolgus macaques to generate in vitro MCM embryos. Time-lapse embryo imaging was performed to assess the timing of MCM embryonic developmental events in control and CRISPR-Cas9 microinjected embryos. Using a dual-guide gene targeting approach, biallelic deletions in the CCR5 gene were introduced into ~ 23-37% of MCM embryos. In addition, single blastomere PCR analysis revealed mosaicism in CCR5 editing within the same embryo. Successful development of IVF and CCR5 editing protocols in MCM embryos lays a foundation for the creation of CCR5-mutant MCMs to assess novel stem cell-based HIV therapeutics.


Subject(s)
Animals, Genetically Modified , CRISPR-Cas Systems , Embryo, Mammalian/metabolism , Gene Editing , Receptors, CCR5 , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Macaca fascicularis , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
8.
J Am Assoc Lab Anim Sci ; 59(6): 681-686, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32878681

ABSTRACT

The objective of this study was to optimize cryopreservation of sperm from Mauritian cynomolgus macaques (MCM) in defined conditions. Sperm viability and motility were compared between sperm cryopreserved in chemically-defined freezing media with variable osmolarity and the presence of either ethylene glycol or glycerol. The highest percentage viability (after freeze-thaw) was seen in sperm samples that were cryopreserved in medium with an osmolarity of 310 mOsm, while higher osmolarities markedly decreased sperm viability. Ethylene glycol and glycerol at concentrations of 4.6% and 5%, respectively, preserved sperm viability to an equivalent degree. Although higher motility rates and higher straight-line velocities were observed in sperm samples frozen in glycerol compared with ethylene glycol, these differences were not statistically significant. Thawed sperm frozen in defined conditions with glycerol were capable of fertilizing MCM oocytes in vitro, with development to the blastocyst stage. The protocol described here provides an effective method for cryopreservation of sperm to facilitate subsequent in vitro fertilization and genome editing of embryos in MCM species.


Subject(s)
Macaca fascicularis , Semen Preservation/veterinary , Animals , Cryopreservation/methods , Cryoprotective Agents/chemistry , Ethylene Glycol/chemistry , Female , Fertilization in Vitro/veterinary , Glycerol/chemistry , Male , Semen Preservation/methods , Spermatozoa/cytology
9.
Sci Rep ; 10(1): 3447, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103062

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) G2019S is a relatively common mutation, associated with 1-3% of Parkinson's disease (PD) cases worldwide. G2019S is hypothesized to increase LRRK2 kinase activity. Dopaminergic neurons derived from induced pluripotent stem cells of PD patients carrying LRRK2 G2019S are reported to have several phenotypes compared to wild type controls, including increased activated caspase-3 and reactive oxygen species (ROS), autophagy dysfunction, and simplification of neurites. The common marmoset is envisioned as a candidate nonhuman primate species for comprehensive modeling of genetic mutations. Here, we report our successful use of CRISPR/Cas9 with repair template-mediated homology directed repair to introduce the LRRK2 G2019S mutation, as well as a truncation of the LRRK2 kinase domain, into marmoset embryonic and induced pluripotent stem cells. We found that, similar to humans, marmoset LRRK2 G2019S resulted in elevated kinase activity. Phenotypic evaluation after dopaminergic differentiation demonstrated LRRK2 G2019S-mediated increased intracellular ROS, decreased neuronal viability, and reduced neurite complexity. Importantly, these phenotypes were not observed in clones with LRRK2 truncation. These results demonstrate the feasibility of inducing monogenic mutations in common marmosets and support the use of this species for generating a novel genetic-based model of PD that expresses physiological levels of LRRK2 G2019S.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease/pathology , Amino Acid Sequence , Animals , Autophagy , Callithrix , Cell Differentiation , Disease Models, Animal , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoplasmic Reticulum Stress , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mutagenesis, Site-Directed , Neurites/physiology , Parkinson Disease/genetics , Phosphorylation , Reactive Oxygen Species/metabolism , Up-Regulation
10.
Biol Reprod ; 102(4): 806-816, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31901091

ABSTRACT

Zika virus (ZIKV) infection is associated with adverse pregnancy outcomes in humans, and infection in the first trimester can lead to miscarriage and stillbirth. Vertical and sexual transmissions of ZIKV have been demonstrated, yet the impact of infection during the initial stages of pregnancy remains unexplored. Here we defined the impact of ZIKV on early embryonic and placental development with a rhesus macaque model. During in vitro fertilization (IVF), macaque gametes were inoculated with a physiologically relevant dose of 5.48log10 plaque-forming units (PFU) of Zika virus/H.sapiens-tc/PUR/2015/PRVABC59_v3c2. Exposure at fertilization did not alter blastocyst formation rates compared to controls. To determine the impact of ZIKV exposure at implantation, hatched blastocysts were incubated with 3.26log10, 4.26log10, or 5.26log10 PFU, or not exposed to ZIKV, followed by extended embryo culture for 10 days. ZIKV exposure negatively impacted attachment, growth, and survival in comparison to controls, with exposure to 5.26log10 PFU ZIKV resulting in embryonic degeneration by day 2. Embryonic secretion of pregnancy hormones was lower in ZIKV-exposed embryos. Increasing levels of infectious virus were detected in the culture media post-exposure, suggesting that the trophectoderm is susceptible to productive ZIKV infection. These results demonstrate that ZIKV exposure severely impacts the zona-free blastocyst, whereas exposure at the time of fertilization does not hinder blastocyst formation. Overall, early stages of pregnancy may be profoundly sensitive to infection and pregnancy loss, and the negative impact of ZIKV infection on pregnancy outcomes may be underestimated.


Subject(s)
Placenta/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/virology , Zika Virus , Animals , Blastocyst/virology , Disease Models, Animal , Female , Fertilization in Vitro , Macaca mulatta , Pregnancy , Trophoblasts/virology
11.
Methods Mol Biol ; 2006: 309-319, 2019.
Article in English | MEDLINE | ID: mdl-31230289

ABSTRACT

The biology of fertility, early development, and pregnancy is variable across mammalian species. In addition, while the physiology and pathophysiology of human diseases can be investigated in other animal models (principally, rodents), differences between human and lower mammals often present limitations in the applicability of physiological processes from rodent models to human biology. Since 1984, when the first live birth from rhesus monkey in vitro fertilization and embryo transfer was reported (Bavister et al., Proc Natl Acad Sci 81:2218-2222, 1984), there has been progress in the implementation of assisted reproductive technologies with several nonhuman primate (NHP) species that play important roles in biomedical research. In recent years, the significance of this progress has been amplified by the development of genomic editing approaches for facile genetic manipulation of the embryo, including methods now applied to NHPs (Liu et al., Cell Stem Cell 14:323-328, 2014; Niu et al., Cell 156:836-843, 2014). In this review, we summarize current protocols and practices for the common marmoset. It is our intention to provide current state-of-the-art protocols for gamete procurement and in vitro fertilization techniques, so that laboratories wishing to implement experimental embryology in marmoset models will have a basic set of tools with which to initiate such studies.


Subject(s)
Embryo Culture Techniques/methods , Embryo Transfer/methods , Embryo, Mammalian/embryology , Fertilization in Vitro/methods , Animals , Callithrix , Embryo, Mammalian/cytology , Female
12.
Placenta ; 65: 55-64, 2018 05.
Article in English | MEDLINE | ID: mdl-29908642

ABSTRACT

OBJECTIVE: Expression of microRNAs (miRNAs) in the human placenta is dynamic across gestation, with expression of miRNAs belonging to the C14MC, C19MC and miR-371-3 clusters. Specifically, miRNAs within the C19MC cluster are exclusively expressed in primates with predominant expression in the placenta. Non-human primates can be utilized to study developmental processes of placentation in vivo that cannot be assessed in the human placenta, however, miRNA expression has not been defined in the macaque placenta. Our objective was to profile miRNAs in the macaque placenta, hypothesizing that expression is conserved between the macaque and human placenta. METHODS: Total RNA from first trimester and term macaque placentas (n = 4 per group) was analyzed through RNA-sequencing and validated by quantitative real-time PCR (qRT-PCR). RESULTS: A total of 607 pre-miRNAs previously annotated in the macaque reference database (miRBase21) were detected, and 166 miRNAs were differentially expressed between first trimester and term placentas. A total of 457 unannotated sequences were detected and deemed candidate novel miRNAs by miRDeep2 software. Differential expression was confirmed for six of nine miRNAs evaluated by qRT-PCR. Comparative analysis demonstrated expression of several miRNA orthologs of human pregnancy-associated miRNA clusters in the macaque placenta. CONCLUSIONS: Profiling placental miRNAs of the macaque revealed conserved expression of a number of miRNAs within the C14MC, C19MC and miR-371-3 clusters between the human and macaque. These results establish non-human primates as a model for human placentation and miRNA biology, with the prediction of their functional significance in placental development and function.


Subject(s)
Macaca mulatta/genetics , MicroRNAs/genetics , Placenta/metabolism , Placentation/genetics , Pregnancy, Animal , Animals , Female , Gene Expression , Gene Expression Profiling/veterinary , Humans , Multigene Family/genetics , Pregnancy , Pregnancy Trimester, First/genetics , Pregnancy Trimester, Third/genetics , Pregnancy, Animal/genetics , Species Specificity , Transcriptome
13.
Reprod Biol Endocrinol ; 16(1): 24, 2018 Mar 16.
Article in English | MEDLINE | ID: mdl-29548332

ABSTRACT

BACKGROUND: The initiation of primate embryo invasion into the endometrium and the formation of the placenta from trophoblasts, fetal mesenchyme, and vascular components are essential for the establishment of a successful pregnancy. The mechanisms which direct morphogenesis of the chorionic villi, and the interactions between trophectoderm-derived trophoblasts and the fetal mesenchyme to direct these processes during placentation are not well understood due to a dearth of systems to examine and manipulate real-time primate implantation. Here we describe an in vitro three-dimensional (3-D) model to study implantation which utilized IVF-generated rhesus monkey embryos cultured in a Matrigel explant system. METHODS: Blastocyst stage embryos were embedded in a 3-D microenvironment of a Matrigel carrier and co-cultured with a feeder layer of cells generating conditioned medium. Throughout the course of embryo co-culture embryo growth and secretions were monitored. Embedded embryos were then sectioned and stained for markers of trophoblast function and differentiation. RESULTS: Signs of implantation were observed including enlargement of the embryo mass, and invasion and proliferation of trophoblast outgrowths. Expression of chorionic gonadotropin defined by immunohistochemical staining, and secretion of chorionic gonadotropin and progesterone coincident with the appearance of trophoblast outgrowths, supported the conclusion that a trophoblast cell lineage formed from implanted embryos. Positive staining for selected markers including Ki67, MHC class I, NeuN, CD31, vonWillebrand Factor and Vimentin, suggest growth and differentiation of the embryo following embedding. CONCLUSIONS: This 3-D in vitro system will facilitate further study of primate embryo biology, with potential to provide a platform for study of genes related to implantation defects and trophoblast differentiation.


Subject(s)
Embryo Implantation/physiology , Macaca mulatta/embryology , Trophoblasts/physiology , Animals , Cell Differentiation , Chorionic Gonadotropin/metabolism , Embryo Culture Techniques/veterinary , Female , Fertilization in Vitro/veterinary , Models, Biological , Morphogenesis , Placentation/physiology , Pregnancy , Progesterone/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...