1
|
Yao H, Sun N, Shao H, Wang T, Tan T. Ex utero embryogenesis of non-human primate embryos and beyond. Curr Opin Genet Dev 2023; 82:102093. [PMID: 37573834 DOI: 10.1016/j.gde.2023.102093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
Understanding cellular and molecular processes underlying the human early post-implantation development represents one of the most fundamental questions in development and stem cell biology. As embryos implant into the uterus a week after fertilization, human development beyond the blastocyst stage is extremely difficult to study due to the inaccessibility of embryos and ethical concerns. The advents in the human embryo in vitro culture system provide an easily accessible, tractable, and perturbable platform to dissect key developmental events of human early embryonic development. However, these studies stopped around gastrulation to technical and ethical limitations, and our understanding of human gastrulation and early organogenesis remains poor. As closely related species to humans, non-human primates (NHPs) are suitable surrogate species to interrogate mechanisms underpinning human embryonic development. Here, we review the most recent advances in embryo in vitro culture systems of NHP and discuss their potential optimization strategies and applications.
Collapse
Affiliation(s)
- Hui Yao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Nianqin Sun
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Honglian Shao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Tianxiang Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Tao Tan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China.
| |
Collapse
|
2
|
Ryu J, Statz JP, Chan W, Burch FC, Brigande JV, Kempton B, Porsov EV, Renner L, McGill T, Burwitz BJ, Hanna CB, Neuringer M, Hennebold JD. CRISPR/Cas9 editing of the MYO7A gene in rhesus macaque embryos to generate a primate model of Usher syndrome type 1B. Sci Rep 2022; 12:10036. [PMID: 35710827 PMCID: PMC9203743 DOI: 10.1038/s41598-022-13689-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022] Open
Abstract
Mutations in the MYO7A gene lead to Usher syndrome type 1B (USH1B), a disease characterized by congenital deafness, vision loss, and balance impairment. To create a nonhuman primate (NHP) USH1B model, CRISPR/Cas9 was used to disrupt MYO7A in rhesus macaque zygotes. The targeting efficiency of Cas9 mRNA and hybridized crRNA-tracrRNA (hyb-gRNA) was compared to Cas9 nuclease (Nuc) protein and synthetic single guide (sg)RNAs. Nuc/sgRNA injection led to higher editing efficiencies relative to mRNA/hyb-gRNAs. Mutations were assessed by preimplantation genetic testing (PGT) and those with the desired mutations were transferred into surrogates. A pregnancy was established from an embryo where 92.1% of the PGT sequencing reads possessed a single G insertion that leads to a premature stop codon. Analysis of single peripheral blood leukocytes from the infant revealed that half the cells possessed the homozygous single base insertion and the remaining cells had the wild-type MYO7A sequence. The infant showed sensitive auditory thresholds beginning at 3 months. Although further optimization is needed, our studies demonstrate that it is feasible to use CRISPR technologies for creating NHP models of human diseases.
Collapse
Affiliation(s)
- Junghyun Ryu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - John P Statz
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA
| | - William Chan
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Fernanda C Burch
- Assisted Reproductive Technologies Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - John V Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Beth Kempton
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Edward V Porsov
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Lauren Renner
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Trevor McGill
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Benjamin J Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Carol B Hanna
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Assisted Reproductive Technologies Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Martha Neuringer
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA.
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
3
|
Chang TA, Bondarenko GI, Gerami-Naini B, Drenzek JG, Durning M, Garthwaite MA, Schmidt JK, Golos TG. Trophoblast differentiation, invasion and hormone secretion in a three-dimensional in vitro implantation model with rhesus monkey embryos. Reprod Biol Endocrinol 2018; 16:24. [PMID: 29548332 PMCID: PMC5857108 DOI: 10.1186/s12958-018-0340-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/04/2018] [Indexed: 11/14/2022] Open
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.
Collapse
Affiliation(s)
- T Arthur Chang
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Gennadiy I Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: Covance Laboratories, Madison, WI, USA
| | - Behzad Gerami-Naini
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: School of Dental Medicine, Tufts University, Boston, MA, USA
| | - Jessica G Drenzek
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: Illumina-Madison, Madison, WI, USA
| | - Maureen Durning
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
| | - Mark A Garthwaite
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
4
|
Romanek J, Opiela J, Smorąg Z. The impact of high hydrostatic pressure (40 MPa and 60 MPa) on the apoptosis rates and functional activity of cryopreserved porcine mesenchymal stem cells. ANNALS OF ANIMAL SCIENCE 2018. [DOI: 10.1515/aoas-2017-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The aim of the present study was to examine the influence of two varied high hydrostatic pressure (HHP) values on the apoptosis (assessing caspase-8, survivin, CAD, Bax, BclxL and BclxS) and functional activity (using cocultures with bovine embryos) of porcine mesenchymal stem cells (pBMSCs). pBMSCs were isolated from porcine bone marrow and cultured in vitro. Before cryopreservation and storage in liquid nitrogen, pBMSCs were subjected to HHP values of 40 MPa and 60 MPa for 1 h at 24°C. After thawing, the cells were analysed for caspase-8 activity and protein expression of survivin, CAD, Bax, BclxL and BclxS. To indirectly test the influence of HHP on the functional activity of pBMSCs, in vitro maturated bovine oocytes were fertilized in vitro, and the obtained embryos were cultured under 4 different conditions: 1. monoculture in SOF medium; 2. coculture with pBMSCs in SOF medium; 3. coculture with pBMSCs subjected to 40 MPa HHP in SOF medium and 4. coculture with pBMSCs subjected to 60 MPa HHP in SOF medium. The quality of the developed blastocysts was analysed by TUNEL assay. HHP did not induce apoptosis in pBMSCs, as no significant difference was noted in the expression of any of the analysed apoptosis- related proteins between pBMSCs subjected to HHP (40 MPa or 60 MPa) and control. The highest number of obtained blastocysts was observed when the embryos were cultured in SOF. A highly significant difference (P<0.005) was noted between embryos cultured in SOF and embryos cultured in the presence of pBMSCs subjected to 60 MPa HHP or untreated pBMSCs. A significant difference (P<0.05) was noted between embryos cultured in SOF and embryos cultured in the presence of pBMSCs subjected to 40 MPa HHP. In conclusion, HHP does not induce apoptosis in pBMSCs. The obtained results of the blastocysts cocultured in vitro with pBMSCs (HHP-treated and untreated cells) imply that coculture with pBMSCs has a negative impact on the developmental rates of blastocysts.
Collapse
Affiliation(s)
- Joanna Romanek
- Department of Biotechnology of Reproduction and Cryopreservation, National Research Institute of Animal Production, 32-083 Balice n. Kraków , Poland
| | - Jolanta Opiela
- Department of Biotechnology of Reproduction and Cryopreservation, National Research Institute of Animal Production, 32-083 Balice n. Kraków , Poland
| | - Zdzisław Smorąg
- Department of Biotechnology of Reproduction and Cryopreservation, National Research Institute of Animal Production, 32-083 Balice n. Kraków , Poland
| |
Collapse
|
5
|
Intracytoplasmic sperm injection into oocytes matured in vitro and early embryonic development in the owl monkey ( Aotus lemurinus). Reprod Med Biol 2015; 15:183-186. [PMID: 29259435 DOI: 10.1007/s12522-015-0229-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/29/2015] [Indexed: 10/22/2022] Open
Abstract
Purpose We explored the possibility of employing intracytoplasmic sperm injection (ICSI), involving oocytes and sperm of owl monkeys, to increase the availability of this species for investigations relating to malaria, etc., by increasing the number of animals in our laboratory. Methods Two owl monkeys (a female and a male), raised at the Amami Laboratory of the University of Tokyo, were used. Follicular oocytes surrounded with cumulus cells were cultured in vitro for approximately 25 h and cumulus cells were removed with 0.1 % hyaluronidase. Because of the poor motility of caudal epididymal sperm, sperm were injected without adding polyvinylpyrrolidone to immobilize them. The ICSI procedure was performed by an individual with considerable experience of human ICSI. Results We were able to produce two owl monkey embryos using ICSI of oocytes that matured to MII stage. Both embryos reached the 10-cell stage at 98 h after ICSI and showed signs of compaction, but failed to cleave further. Conclusions Although we successfully produced owl monkey embryos after ICSI, the embryos did not develop to the blastocyst stage. Many parameters need to be studied further, including superovulation, selection of culture media, and selection of good quality sperm in order to achieve successful ICSI in the owl monkey.
Collapse
|
6
|
Abstract
Nonhuman primates (NHP) are the closest animal species to humans and have been widely used for studying human reproductive physiology. Assisted reproductive technology (ART) in Old World NHPs provides great opportunity for studying fertilization, embryo development, embryonic stem cell (ESC) derivation for regenerative medicine, somatic cell nuclear transfer (cloning), and transgenic NHP models of inherited genetic disorders. Here we present two ART protocols developed for rhesus monkey (Macaca mulatta) and baboon (Papio cynocephalus).
Collapse
Affiliation(s)
- Tien-Cheng Arthur Chang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| | | |
Collapse
|
7
|
Sparman ML, Tachibana M, Mitalipov SM. Cloning of non-human primates: the road "less traveled by". THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2011; 54:1671-8. [PMID: 21404187 DOI: 10.1387/ijdb.103196ms] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Early studies on cloning of non-human primates by nuclear transfer utilized embryonic blastomeres from preimplantation embryos which resulted in the reproducible birth of live offspring. Soon after, the focus shifted to employing somatic cells as a source of donor nuclei (somatic cell nuclear transfer, SCNT). However, initial efforts were plagued with inefficient nuclear reprogramming and poor embryonic development when standard SCNT methods were utilized. Implementation of several key SCNT modifications was critical to overcome these problems. In particular, a non-invasive method of visualizing the metaphase chromosomes during enucleation was developed to preserve the reprogramming capacity of monkey oocytes. These modifications dramatically improved the efficiency of SCNT, yielding high blastocyst development in vitro. To date, SCNT has been successfully used to derive pluripotent embryonic stem cells (ESCs) from adult monkey skin fibroblasts. These remarkable advances have the potential for development of human autologous ESCs and cures for many human diseases. Reproductive cloning of nonhuman primates by SCNT has not been achieved yet. We have been able to establish several pregnancies with SCNT embryos which, so far, did not progress to term. In this review, we summarize the approaches, obstacles and accomplishments of SCNT in a non-human primate model.
Collapse
Affiliation(s)
- Michelle L Sparman
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | | | | |
Collapse
|
8
|
Vandevoort CA, Mtango NR, Latham KE, Stewart DR. Primate preimplantation embryo is a target for relaxin during early pregnancy. Fertil Steril 2011; 96:203-7. [PMID: 21645893 DOI: 10.1016/j.fertnstert.2011.05.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To determine whether preimplantation embryos are targets for relaxin secreted from the corpus luteum of the menstrual cycle. DESIGN Rhesus monkey oocytes obtained from females undergoing controlled ovarian hyperstimulation were inseminated, and the resulting embryos were cultured in medium with or without recombinant human relaxin (20 ng/mL) for 8 days. SETTING Research laboratory. ANIMAL(S) Rhesus monkey. INTERVENTION(S) Controlled ovarian stimulation to obtain oocytes for in vitro-produced embryos that were cultured with or without human recombinant relaxin. MAIN OUTCOME MEASURE(S) Rate of blastocyst development, percentage of blastocysts, and inner cell mass/trophectoderm cell ratio were measured on day 8 of culture. The presence of relaxin receptor (RXFP1) messenger RNA in eight-cell embryos was observed by array hybridization. RESULT(S) RXFP1 receptor expression was localized to the inner cell mass of blastocysts, as shown by immunohistochemistry. The percentage of embryos that developed to blastocyst and the inner cell mass/trophectoderm cell ratio was unchanged with relaxin supplementation; however, the relaxin-treated embryos developed into blastocysts significantly sooner than untreated embryos. CONCLUSION(S) These results are the first evidence that the preimplantation primate embryo is a target for relaxin and that the addition of relaxin to in vitro culture medium enhances rhesus monkey embryo development.
Collapse
Affiliation(s)
- Catherine A Vandevoort
- California National Primate Research Center, University of California, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
9
|
Klooster KL, Burruel VR, Meyers SA. Loss of fertilization potential of desiccated rhesus macaque spermatozoa following prolonged storage. Cryobiology 2011; 62:161-6. [DOI: 10.1016/j.cryobiol.2011.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/03/2011] [Accepted: 02/04/2011] [Indexed: 10/18/2022]
|
10
|
Chang TC, Eddy CA, Ying Y, Liu YG, Holden AE, Brzyski RG, Schenken RS. Ovarian stimulation, in vitro fertilization, and effects of culture conditions on baboon preimplantation embryo development. Fertil Steril 2011; 95:1217-23. [DOI: 10.1016/j.fertnstert.2010.06.095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
|
11
|
Shimozawa N, Nakamura S, Takahashi I, Hatori M, Sankai T. Characterization of a novel embryonic stem cell line from an ICSI-derived blastocyst in the African green monkey. Reproduction 2010; 139:565-73. [DOI: 10.1530/rep-09-0067] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several cell types from the African green monkey (Cercopithecus aethiops), such as red blood cells, primary culture cells from kidney, and the Vero cell line, are valuable sources for biomedical research and testing. Embryonic stem (ES) cells that are established from blastocysts have pluripotency to differentiate into these and other types of cells. We examined an in vitro culture system of zygotes produced by ICSI in African green monkeys and attempted to establish ES cells. Culturing with and without a mouse embryonic fibroblast (MEF) cell monolayer resulted in the development of ICSI-derived zygotes to the blastocyst stage, while culturing with a buffalo rat liver cell monolayer yielded no development (3/14, 21.4% and 6/31, 19.4% vs 0/23, 0% respectively; P<0.05). One of the nine blastocysts, which had been one of the zygotes co-cultured with MEF cells, formed flat colonies consisting of cells with large nuclei, similar to other primate ES cell lines. The African green monkey ES (AgMES) cells expressed pluripotency markers, formed teratomas consisting of three embryonic germ layer tissues, and had a normal chromosome number. Furthermore, expression of the germ cell markers CD9 and DPPA3 (STELLA) was detected in the embryoid bodies, suggesting that AgMES cells might have the potential ability to differentiate into germ cells. The results suggested that MEF cells greatly affected the quality of the inner cell mass of the blastocysts. In addition, AgMES cells would be a precious resource for biomedical research such as other primate ES cell lines.
Collapse
|
12
|
Kubisch HM, Gagliardi C, Romero DG, Bunnell BA, Ratterree MS. Kinetics of pronuclear development and the effects of vector type and timing of injection on the efficiency of gene transfer into rhesus macaque embryos. Mol Reprod Dev 2008; 75:1505-14. [PMID: 18361395 DOI: 10.1002/mrd.20901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A series of experiments was performed to determine the dynamics of pronuclear development as well as the efficiency of either adenovirus-associated (AAV) or lentivirus-derived vectors to introduce a green fluorescent protein (GFP) reporter gene into rhesus macaque (Macaca mulatta) embryos. Assessment of pronuclear development at various times after fertilization revealed that the appearance of pronuclei was determined by the presence of the first and the timing of the second polar body. The dynamics of pronuclear formation was a significant determinant of whether an oocyte reached the blastocyst stage, however, when the percentage of blastocysts were based on the number of zygotes, the timing of the appearance of polar bodies did not appear to have any effect on subsequent development. Injection of different AAV-derived vectors showed that the serotype of the vector did not affect development or the proportion of transgenic embryos. Moreover, all putative transgenic embryos proved to be expression mosaics. Injection of embryos with lentiviral vectors showed that timing of injection (before or after fertilization) had no effect on subsequent transgene expression, but that the type of reporter gene determined post-injection development and rate of transgenesis. The transfer of embryos following injection of a lentiviral vector into three recipients resulted in one pregnancy which was lost during the second trimester. Analysis of fetal tissues showed ubiquitous presence of the transgene and GFP expression in all tissues examined. These results show that lentivirus-derived vectors can efficiently transform rhesus embryos and are suitable for the generation of transgenic rhesus monkeys.
Collapse
Affiliation(s)
- H M Kubisch
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.
| | | | | | | | | |
Collapse
|
13
|
Sun Q, Dong J, Yang W, Jin Y, Yang M, Wang Y, Wang PL, Hu Y, Tsien JZ. Efficient reproduction of cynomolgus monkey using pronuclear embryo transfer technique. Proc Natl Acad Sci U S A 2008; 105:12956-60. [PMID: 18725640 PMCID: PMC2529107 DOI: 10.1073/pnas.0805639105] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Indexed: 11/18/2022] Open
Abstract
One of the technical bottlenecks in producing nonhuman primate models is that current assisted reproductive techniques, such as in vitro culture and frozen conservation of multicell-stage embryos, often result in poor embryo quality and subsequently lead to low birth rates. We investigated whether pronuclear embryo transfer can be used as an effective means for improving pregnancy and live birth rates of nonhuman primates. We collected 174 metaphase II oocytes by laparoscopy from 22 superovulated mature females and then fertilized these eggs using either in vitro fertilization or intracytoplasmic sperm injection, resulting in a 33.3% and a 50% fertilization rate, respectively. These 66 fertilized pronuclear-stage embryos were then tubally transferred to 30 recipients and led to 7 births and 1 abortion. Importantly, we observed that the highest live birth rate of approximately 64% was obtained when the transfer of pronuclear embryos was performed in the presence of new corpus luteum in the ovary of recipients between 24 h and 36 h after estradiol peak. Therefore, our experiments demonstrate that by matching the critical time window in the recipient's reproductive cycle for achieving optimal embryo-uterine synchrony, pronuclear embryo transfer technology can significantly improve the pregnancy rate and live birth of healthy baby monkeys. This efficient method should be valuable to the systematic efforts in construction of various transgenic primate disease models.
Collapse
Affiliation(s)
- Qiang Sun
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
| | - Juan Dong
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| | - Wenting Yang
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| | - Yujuan Jin
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
| | - Mingying Yang
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
| | - Yan Wang
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
| | - Philip L. Wang
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| | - Yinghe Hu
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
| | - Joe Z. Tsien
- *Yunnan Banna Primate Disease Model Research Center, Shanghai Institute of Brain Functional Genomics, Key Lab of Brain Functional Genomics, Ministry of Education and Science and Technology Commission of Shanghai Municipality, East China Normal University, Shanghai 200062, People's Republic of China; and
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| |
Collapse
|
14
|
Yeoman RR, Mitalipov S, Gerami-Naini B, Nusser KD, Wolf DP. Low temperature storage of rhesus monkey spermatozoa and fertility evaluation by intracytoplasmic injection. Theriogenology 2005; 63:2356-71. [PMID: 15910919 DOI: 10.1016/j.theriogenology.2004.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2004] [Accepted: 05/11/2004] [Indexed: 11/22/2022]
Abstract
The objective was to develop a sperm freezing procedure suitable for use in the propagation of valuable founder animals by assisted reproductive technologies. Here, we report a comparison of processing methods by measuring the motility of fresh and frozen-thawed rhesus monkey spermatozoa and fertility via intracytoplasmic spermatozoa injection (ICSI) of sibling oocytes. Washed spermatozoa were frozen in straws or in pellets using different cryoprotective media and processed post-thaw with or without a density gradient centrifugation step. Among the four study series, motility post-thaw was improved with density gradient centrifugation (17-24% versus 75%, P<0.01) achieving levels similar to fresh spermatozoa. Spermatozoa injected oocytes (total n=377) were co-cultured on BRL cells and observed for fertilization and development. With spermatozoa frozen in straws in liquid nitrogen vapors, the fertilization rate after ICSI was lower than with fresh spermatozoa (40-44% versus 77-86%, P<0.05), even with the Percoll-enriched fraction that exhibited robust motility. In contrast, somewhat slower freezing of spermatozoa in pellets on dry ice supported fertilization rates (73%) that were similar to the fresh counterpart. Developmental rates of fertilized eggs were similar in all experiments. A total of 106 embryo transfers has resulted in the first primate born after ICSI with F/T ejaculated spermatozoa plus 22 other infants to date. Additionally, a 3-4 h incubation after thawing improved the fertilization rate with spermatozoa from a male with poor post-thaw recovery of sperm motility. In conclusion, an acceptable fertilization rate after ICSI with motile, frozen-thawed primate spermatozoa was observed comparable to that obtained with fresh spermatozoa allowing small quantities of competent spermatozoa to be used with ICSI to facilitate propagation of desirable primate genotypes.
Collapse
Affiliation(s)
- Richard R Yeoman
- Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health and Science University, 505 185th Avenue, Beaverton, Oregon, OR 97006, USA.
| | | | | | | | | |
Collapse
|
15
|
Enders AC, Meyers S, Vandevoort CA, Douglas GC. Interactions of macaque blastocysts with epithelial cells in vitro. Hum Reprod 2005; 20:3026-32. [PMID: 16006466 DOI: 10.1093/humrep/dei178] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Early in vitro studies of blastocyst formation in several primate species have demonstrated the feasibility of such studies. Initial studies of in vitro-fertilized oocytes cultured with buffalo rat liver cells suggested that other epithelial cells might be used to assess blastocyst adherence and penetration in vitro. METHODS Macaque blastocysts were incubated with different epithelial cell lines or with Matrigel. The interaction was studied using light and transmission electron microscopy. RESULTS In general, zona-free blastocysts attached 2 days after placing on the substrates. MDCK cells provided optimal conditions for blastocyst development. The best preparations showed some development of an amniotic cavity and distribution of cytotrophoblast and syncytial trophoblast. Distribution of syncytial trophoblast at the margin of the site and cytotrophoblast centrally was similar to that seen at the trophoblastic plate stage in this species. However, there was less syncytial trophoblast than is normally found at this stage, and total time from fertilization to the trophoblastic plate stage was delayed 2 days. CONCLUSIONS While in vitro studies with blastocysts cannot completely mimic the intrauterine environment, they can illustrate some of the potential interactions and provide a situation in which parameters may be manipulated.
Collapse
Affiliation(s)
- Allen C Enders
- Department of Cell Biology and Human Anatomy, University of California, Davis, California 95616, USA.
| | | | | | | |
Collapse
|
16
|
Okada H, Hirose Y, Manonmani P, Uda A, Ito M, Sankai T. Characterization of an immortalized oviduct cell line from the cynomolgus monkey (Macaca fascicularis). J Med Primatol 2005; 34:67-72. [PMID: 15860112 DOI: 10.1111/j.1600-0684.2005.00093.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To establish reproductive biological techniques in mammals, it is important to understand the growth environment of the embryo. Oviduct epithelial cells are in close proximity to the embryo during pre-implantation development. We, therefore, established an immortalized oviduct epithelial cell line from the cynomolgus monkey, evaluated the usefulness of these cells as feeder cells for embryo culture, and investigated the gene expression of several growth factors and cytokines in the cells. The immortalized cells were positive for the anti-cytokeratin antibody, as determined by immunocytochemistry, indicating that they are epithelial. They also expressed oviductin, which is specific to oviduct epithelial cells, glyceraldehyde-3-phosphate dehydrogenase (control), leukemia inhibitory factor, vascular endothelial growth factor, epidermal growth factor, insulin-like growth factor 1, transforming growth factor beta-2, and interleukin 4. Mouse embryo development was improved when the immortalized cells were used as feeder cells. This cell line is also useful for studying the factors secreted by oviduct epithelial cells.
Collapse
Affiliation(s)
- H Okada
- Tsukuba Primate Center for Medical Science, National Institute of Infectious Diseases, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
The assisted reproductive technologies (ARTs) have been used in the production of rhesus monkey offspring at the Oregon National Primate Research Center (ONPRC) and that experience is summarized here. Additionally these technologies serve as a source of oocytes/embryos for monozygotic twinning, embryonic stem (ES) cell derivation and cloning. High fertilization efficiencies were realized with conventional insemination or following the use of intracytoplasmic sperm injection (ICSI) and approximately 50% of the resulting embryos grew in vitro to blastocysts. Both fresh and frozen sperm were employed in fertilization by ICSI and the resulting embryos could be low temperature stored for subsequent thawing and transfer when a synchronized recipient female was available or after shipment to another facility. Following the transfer of up to 3 embryos, an overall pregnancy rate of 30% was achieved with increasing rates dependent upon the number of embryos transferred. Singleton pregnancy outcomes following the transfer of ART produced embryos were similar to those observed in a control group of animals in the timed mated breeding colony at ONPRC. ICSI produced embryos were used in efforts to create monozygotic twins by blastomere separation or blastocyst splitting. While pregnancies were achieved following the transfer of demi-embryos, only one was a twin and it was lost to spontaneous abortion. ICSI produced embryos have also served as the source of blastocysts for the derivation of embryonic stem cells. These pluripotent cells hold potential for cell based therapies and we consider the monkey an important translational model in which to evaluate safety, efficacy and feasibility of regenerative medicine approaches based on the transplantation of stem cell-derived progeny. Finally, efforts to produce genetically-identical monkeys by nuclear transfer have been briefly summarized.
Collapse
Affiliation(s)
- Don P Wolf
- Division of Reproductive Sciences, Oregon National Primate Research Center Beaverton, Oregon, USA.
| |
Collapse
|
18
|
Wolf DP, Thormahlen S, Ramsey C, Yeoman RR, Fanton J, Mitalipov S. Use of assisted reproductive technologies in the propagation of rhesus macaque offspring. Biol Reprod 2004; 71:486-93. [PMID: 15044263 DOI: 10.1095/biolreprod.103.025932] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The assisted reproductive technologies (ARTs) as tailored to the production of rhesus monkeys at the Oregon National Primate Research Center (ONPRC) are described. Efficient fertilization of mature oocytes recovered by aspiration from females subjected to follicular stimulation was achieved with fresh or frozen sperm by intracytoplasmic sperm injection (ICSI). Embryo development to the early cleavage stage occurred at high frequency. Cryopreserved embryos showed high postthaw survival and were also transferred in efforts to establish pregnancies. Three methods of transfer were evaluated, two involving embryo placement into the oviduct, laparoscopy and minilaparotomy, and a nonsurgical, transcervical approach that resulted in uterine deposition. Early cleaving embryos (Days 1-4) were transferred into the oviducts of synchronized recipients with optimal results and pregnancy rates of up to 36%. Pregnancy rates were similar when two fresh or frozen embryos were transferred (28- 30%), although more than two embryos had to be thawed to compensate for embryo loss during freeze-thawing. Normal gestational lengths, birth weights, and growth curves were seen with ART-produced infants compared with infants produced by natural mating in the timed mated breeding (TMB) colony at the ONPRC. In 72 singleton pregnancies established following the transfer of ART-produced embryos, the live-birth rate, at 87.5%, was statistically identical to that for the TMB colony. Further development of the ARTs should result in increasing use of these techniques to augment conventional approaches to propagating monkeys, especially those of defined genotypes.
Collapse
Affiliation(s)
- D P Wolf
- Division of Reproductive Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Borman SM, Chwalisz K, Stouffer RL, Zelinski-Wooten MB. Chronic low-dose antiprogestin impairs preimplantation embryogenesis, but not oocyte nuclear maturation or fertilization in rhesus monkeys. Steroids 2003; 68:1041-51. [PMID: 14667997 DOI: 10.1016/s0039-128x(03)00143-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Continual administration of low doses of the antiprogestin ZK137316 permits ovarian/menstrual cyclicity, but prevents pregnancy in female rhesus monkeys. The sites of contraceptive action remain unknown. This study determined whether chronic, low-dose antiprogestin exposure during follicular development impairs oocyte maturation in vivo, as well as fertilization and preimplantation embryogenesis in vitro. Adult, female rhesus monkeys exhibiting normal menstrual cycles received vehicle (n=9) or 0.03 mg ZK137316 (n=8)/kg body weight i.m. daily for 3 months. Controlled ovarian stimulation with recombinant gonadotropins was initiated in the 3rd month. Oocytes collected from preovulatory follicles were evaluated for nuclear maturity and inseminated in vitro. Preimplantation embryonic development was monitored in vitro. The total number of oocytes and percentage collected at each nuclear stage were similar in both groups. More (P<0.05) atretic oocytes were recovered following antiprogestin relative to vehicle treatment. Fertilization rates and percentages of embryos that progressed to the morula stage were similar between groups, but antiprogestin-treated females exhibited less (P<0.05) normal cleavage. Embryonic development was accelerated by 1 day (P<0.05) from the 16-cell to the morula stage in the antiprogestin group relative to vehicle. Despite this, the majority of embryos became blastocysts within 6 days in vitro in the antiprogestin group, but fewer expanded (P=0.09) and hatched (P<0.05) compared to vehicle. During in vivo treatment with chronic, low-dose antiprogestin, oocytes retained their ability to resume and complete meiosis as well as fertilize following insemination in vitro. However, preimplantation embryogenesis in vitro was impaired, particularly during the later stages of blastocyst development. Thus, antiprogestin exposure during follicular development altered oocyte functions that are critical for normal preimplantation embryogenesis; this may contribute to pregnancy prevention.
Collapse
Affiliation(s)
- Sherri M Borman
- Division of Reproductive Sciences, Oregon National Primate Research Center, 505 N.W. 185th Avenue, Beaverton, OR 97006, USA
| | | | | | | |
Collapse
|
20
|
Curnow EC, Pawitri D, Hayes ES. Sequential culture medium promotes the in vitro development of Macaca fascicularis embryos to blastocysts. Am J Primatol 2002; 57:203-12. [PMID: 12210672 DOI: 10.1002/ajp.10043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In vitro production of blastocyst stage embryos from Macaca fascicularis (Mf) has not previously been demonstrated without cell support. Historical data indicates that a large proportion of Mf embryos arrest at the morula stage in nonsequential culture medium (NSM) lacking serum supplementation and/or cell support. Here we report the application of a sequential culture system supporting in vitro production of Mf blastocysts. Mf embryos produced by in vitro fertilization (IVF; n = 69) were subjected to in vitro culture without cell support in either a commercial sequential embryo culture medium (SM) or an NSM. At 24 hr post-insemination (PI) embryos generated from in vivo and in vitro matured oocytes and cultured in the NSM cleaved to two or more cells in significantly greater proportions (15/23; 65%) compared to embryos cultured in SM (14/46; 30%). However, by day 3 PI embryo development beyond eight cells was not different in NSM (9/23; 39%) compared to SM (25/46; 54%). At day 5 PI embryo development to the morula stage was slightly lower in NSM (8/23, 35%) compared to SM (21/46, 45%), and embryo degeneration was slightly higher in NSM (9/23, 39%) compared to SM (9/46, 20%). After 7-9 days of in vitro culture, embryo development to the blastocyst stage and embryo degeneration were significantly lower and higher, respectively, in NSM (0/23, 0%; and 23/23, 100%) compared to SM (9/46, 20%; and 26/46, 56%). In this study the sequential culture system was better able to support in vitro development of Mf embryos compared to nonsequential culture systems.
Collapse
Affiliation(s)
- E C Curnow
- Monash Institute of Reproduction and Development, Clayton, Australia
| | | | | |
Collapse
|
21
|
Yeoman RR, Gerami-Naini B, Mitalipov S, Nusser KD, Widmann-Browning AA, Wolf DP. Cryoloop vitrification yields superior survival of Rhesus monkey blastocysts. Hum Reprod 2001; 16:1965-9. [PMID: 11527906 DOI: 10.1093/humrep/16.9.1965] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Vitrification using the cryoloop procedure was evaluated for preservation of non-human primate blastocysts by comparing survival results from two different cryoprotectant mixtures with prior results from controlled rate cooling. METHODS Rhesus monkey blastocysts were produced by intracytoplasmic sperm injection of mature oocytes from cycling females stimulated with recombinant human hormones. Morphologically well-formed blastocysts were divided between Procedure A (2.8 mol/l dimethylsulphoxide and 3.6 mol/l ethylene glycol with 0.65 mol/l sucrose and 25 micromol/l Ficoll in TALP-HEPES with 20% fetal bovine serum (TH20)) and Procedure B (3.4 mol/l glycerol and 4.5 mol/l ethylene glycol in TH20). After >48 h in liquid nitrogen, the removal of cryoprotectants was accomplished in the presence of a 3-step series of decreasing sucrose concentrations in TH20. Surviving embryos were co-cultured on buffalo rat liver cells. RESULTS Of 16 blastocysts vitrified via Procedure A, 38% survived with minimal lysis and only one hatched in culture; in contrast, of 33 blastocysts vitrified by Procedure B, 85% survived and 71% hatched. Of 22 blastocysts cryopreserved by conventional slow cooling, 36% survived and 6% hatched. Transfer into three recipients, each with two embryos vitrified with Procedure B, resulted in a successful twin-term pregnancy. CONCLUSION Modified cryoloop vitrification with a final solution of 3.4 mol/l glycerol and 4.5 mol/l ethylene glycol is a promising procedure for preserving Rhesus monkey blastocysts that is simple, rapid, and inexpensive.
Collapse
Affiliation(s)
- R R Yeoman
- Andrology/Embryology Laboratory, Department of Obstetrics/Gynecology, Oregon Health Sciences University, Portland, Oregon 97201, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Nusser KD, Mitalipov S, Widmann A, Gerami-Naini B, Yeoman RR, Wolf DP. Developmental competence of oocytes after ICSI in the rhesus monkey. Hum Reprod 2001; 16:130-137. [PMID: 11139551 DOI: 10.1093/humrep/16.1.130] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oocyte quantity and quality are critical to assisted reproductive technology (ART), yet few assessments beyond counting metaphase II (MII) oocytes exist. In this study, 30 +/- 2 oocytes per cycle were recovered from rhesus monkeys subjected to follicular stimulation with human gonadotrophins, of which 15 +/- 1 were MII. Oocyte quality was investigated by monitoring the developmental potential of oocytes subjected to intracytoplasmic sperm injection (ICSI). Despite uniform fertilization rates (71 +/- 4%), progression of embryos to blastocysts varied when expressed as a monthly average, from 20 to 85%, with lows from February to April and again in October, which could be attributed to developmental failure of a significant number of oocyte cohorts (14 of 55). Blastocyst rates, after elimination of failed cohorts, were uniform over time (59 +/- 4%). Neither culture conditions, the number of follicular stimulations, nor the individual sperm or oocyte donor were associated specifically with developmental failure, suggesting that intrinsic differences between stimulation cycles account for the observed variation in developmental potential. The in-vivo developmental competence of ICSI-produced embryos grown to blastocysts in vitro was also assessed. Two ongoing pregnancies and the birth of a normal female, 'Blastulina', represent landmarks in efforts to expand the use of ART in the rhesus monkey.
Collapse
Affiliation(s)
- K D Nusser
- Division of Reproductive Sciences, Oregon Regional Primate Research Center, Beaverton, OR 97006-3348 and Departments of Obstetrics and Gynecology and Physiology and Pharmacology, Oregon Health Sciences University, Portland, OR 97201-3098, USA
| | | | | | | | | | | |
Collapse
|
23
|
Sankai T. In vitro manipulation of nonhuman primate gametes for embryo production and embryo transfer. Exp Anim 2000; 49:69-81. [PMID: 10889945 DOI: 10.1538/expanim.49.69] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Since nonhuman primates are closely related to humans and share many physical similarities, they are important for use in research areas such as human infectious diseases, reproduction, physiology, endocrinology, metabolism, neurology and longevity. To develop and maintain these animals, we must establish techniques for in vitro manipulation of spermatozoa and eggs. For a decade my research group has been conducting basic research to establish embryo manipulation techniques and to clarify the reproductive phenomena in nonhuman primates. This article summarizes the past research on in vitro manipulation of nonhuman primate gametes, from collection of reproductive cells and in vitro fertilization to the birth of offspring after embryo transfer, as well as the current status of these research areas. The studies summarized here will directly lead to the development of standard techniques for practical and comprehensive use in nonhuman primates.
Collapse
Affiliation(s)
- T Sankai
- Tsukuba Primate Center, National Institute of Infectious Diseases, Ibaraki, Japan
| |
Collapse
|
24
|
Wolf DP, Meng L, Ouhibi N, Zelinski-Wooten M. Nuclear transfer in the rhesus monkey: practical and basic implications. Biol Reprod 1999; 60:199-204. [PMID: 9915982 DOI: 10.1095/biolreprod60.2.199] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In early 1997, the birth of a lamb after transfer of the nucleus from an adult mammary gland cell into an enucleated oocyte, along with the production of rhesus monkeys by nuclear transfer of embryonic cells, marked a reemergence of the field of mammalian cloning. Clonally derived rhesus monkeys would be invaluable in biomedical research, and the commercial interests in transgenic sheep and cattle propagated by cloning are substantial. Nuclear transfer technology is under consideration in human in vitro fertilization clinics to overcome infertility secondary to advanced maternal age or mitochondrial-based genetic disease. Nuclear transfer involves preparing a cytoplast as a recipient cell, in most cases a mature metaphase II oocyte from which the chromosomes have been removed. A donor nucleus cell is then placed between the zona and the cytoplast, and fusion, as well as cytoplast activation, is initiated by electrical stimulation. Successful reprogramming of the donor cell nucleus by the cytoplast is critical--a step that may be influenced by cell cycle stage. Embryos produced by nuclear transfer are cultured in vitro for several cell divisions before cryopreservation or transfer to the oviduct or uterus of a host mother. The efficiency of producing live young by nuclear transfer in domestic species is low, with a high frequency of developmental abnormalities in both preterm and term animals. However, a number of pregnancies have now been established using fetal cells as the source of donor nuclei. The use of cell lines not only allows large clone sizes but also supports the ability to genetically manipulate cells in vitro before nuclear transfer. Ongoing research focused on the production of clonally derived rhesus monkeys using fetal fibroblasts and embryonic stem cells as the source of donor nuclei will be reviewed.
Collapse
Affiliation(s)
- D P Wolf
- Division of Reproductive Sciences, Oregon Regional Primate Research Center, Beaverton, Oregon 97006, USA.
| | | | | | | |
Collapse
|
25
|
Abstract
Primate embryonic stem (ES) cells are derived from preimplantation embryos, have a normal karyotype, and are capable of indefinite, undifferentiated proliferation. Even after culture for more than a year, primate ES cells maintain the potential to differentiate to trophoblast and derivatives of embryonic endoderm, mesoderm, and ectoderm. In this review, we compare the characteristics of ES cell lines from two primate species, the rhesus monkey (Macaca mulatta) and the common marmoset (Callithrix jacchus), with the characteristics of mouse ES cells and human embryonal carcinoma cells. We also discuss the implications of using primate ES cells to understand early human development and discuss the practical and ethical implications for the understanding and treatment of human disease.
Collapse
Affiliation(s)
- J A Thomson
- Wisconsin Regional Primate Research Center, University of Wisconsin, Madison 53715-1299, USA
| | | |
Collapse
|
26
|
Abstract
Several media, some augmented with amino acids, have been formulated recently, based on simplex optimization, to support the preimplantation development of mouse embryos. For the highly limited studies on preimplantation development of nonhuman primate embryos, a complex medium (CMRL-1066) has been employed. Our objective was to compare the developmental ability of rhesus monkey embryos in a simple medium containing amino acids, KSOM/AA, with the complex media used previously. Zygotes (99) were recovered following in vitro fertilization (IVF) from six monkeys, allocated to either CMRL or KSOM/AA both containing 10% fetal calf serum (FCS), and monitored daily until reaching the expanded or hatched blastocyst stage. The distribution of cells between the inner cell mass (ICM) and trophectoderm was determined at the end of culture by differential nuclear staining. Although a greater number of embryos cultured in KSOM/AA vs. CMRL developed to the morula stage (80%) and beyond (66% to expanded blastocyst), the differences were not significant. Such embryos in KSOM/AA did, however, develop at a significantly faster rate, on average, reaching the expanded blastocyst stage 26 hr earlier than did embryos cultured in CMRL. KSOM/AA embryos hatched in less time and had a higher percentage (43 vs. 34) of cells allocated to the ICM. These results indicate that a simple medium, KSOM/AA, in the presence of serum, supports the development of rhesus monkey embryos at high efficiency and at a faster rate than that observed for embryos cultured in the complex medium, CMRL-1066.
Collapse
Affiliation(s)
- A M Weston
- Division of Reproductive Sciences, Oregon Regional Primate Research Center, Beaverton 97006, USA
| | | |
Collapse
|
27
|
Schramm RD, Bavister BD. Use of purified porcine follicle-stimulating hormone for ovarian stimulation of macaque monkeys. Theriogenology 1996; 45:727-32. [PMID: 16727835 DOI: 10.1016/0093-691x(96)00002-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/1995] [Accepted: 09/15/1995] [Indexed: 11/29/2022]
Abstract
At present, in nonhuman primates, ovarian stimulation with heterologous gonadotropin preparations is the only reliable way to produce substantial numbers of competent ova for in vitro fertilization and embryo development studies. Preparations such as equine chorionic gonadotropin (eCG) and human menopausal gonadotropins (hMG or hFSH) have been used successfully, but eCG is crude and contains variable amounts of LH activity, while hMG/hFSH is very expensive and the supply is not stable. This study examined the use of a purified porcine FSH preparation (Folltropin V) for ovarian stimulation in rhesus monkeys. Twice-daily intramuscular injections of this preparation resulted in good follicular development, and was followed by a single intramuscular injection of hCG. Ova were collected laparoscopically 30 h post hCG, fertilized in vitro and then cultured until development ceased. Stimulation of 9 monkeys with Folltropin V yielded a mean of 20 ova per animal, of which 71% reached metaphase II and were inseminated; of these, 92% were fertilized in vitro and 48% developed into blastocysts in vitro. These results are similar to those reported by us and by others using eCG, hMG or an hFSH/hMG combination for ovarian stimulation of macaque monkeys. We conclude that Folltropin V is a suitable alternative preparation for ovarian stimulation in nonhuman primates and one that also has the advantages of being readily available and much less expensive than human gonadotropin preparations.
Collapse
Affiliation(s)
- R D Schramm
- University of Wisconsin, Regional Primate Research Center, 1223 Capitol Court, Madison, WI 53715, USA
| | | |
Collapse
|