Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 511(7508): 177-83, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25008523

ABSTRACT

Human pluripotent stem cells hold potential for regenerative medicine, but available cell types have significant limitations. Although embryonic stem cells (ES cells) from in vitro fertilized embryos (IVF ES cells) represent the 'gold standard', they are allogeneic to patients. Autologous induced pluripotent stem cells (iPS cells) are prone to epigenetic and transcriptional aberrations. To determine whether such abnormalities are intrinsic to somatic cell reprogramming or secondary to the reprogramming method, genetically matched sets of human IVF ES cells, iPS cells and nuclear transfer ES cells (NT ES cells) derived by somatic cell nuclear transfer (SCNT) were subjected to genome-wide analyses. Both NT ES cells and iPS cells derived from the same somatic cells contained comparable numbers of de novo copy number variations. In contrast, DNA methylation and transcriptome profiles of NT ES cells corresponded closely to those of IVF ES cells, whereas iPS cells differed and retained residual DNA methylation patterns typical of parental somatic cells. Thus, human somatic cells can be faithfully reprogrammed to pluripotency by SCNT and are therefore ideal for cell replacement therapies.


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells/metabolism , Animals , Cell Line , Chromosome Aberrations , Chromosomes, Human, X/genetics , Chromosomes, Human, X/metabolism , DNA Copy Number Variations , DNA Methylation , Genome-Wide Association Study , Genomic Imprinting , Humans , Nuclear Transfer Techniques/standards , Pluripotent Stem Cells/cytology , Transcriptome
2.
Nature ; 509(7498): 101-4, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24670652

ABSTRACT

Successful mammalian cloning using somatic cell nuclear transfer (SCNT) into unfertilized, metaphase II (MII)-arrested oocytes attests to the cytoplasmic presence of reprogramming factors capable of inducing totipotency in somatic cell nuclei. However, these poorly defined maternal factors presumably decline sharply after fertilization, as the cytoplasm of pronuclear-stage zygotes is reportedly inactive. Recent evidence suggests that zygotic cytoplasm, if maintained at metaphase, can also support derivation of embryonic stem (ES) cells after SCNT, albeit at low efficiency. This led to the conclusion that critical oocyte reprogramming factors present in the metaphase but not in the interphase cytoplasm are 'trapped' inside the nucleus during interphase and effectively removed during enucleation. Here we investigated the presence of reprogramming activity in the cytoplasm of interphase two-cell mouse embryos (I2C). First, the presence of candidate reprogramming factors was documented in both intact and enucleated metaphase and interphase zygotes and two-cell embryos. Consequently, enucleation did not provide a likely explanation for the inability of interphase cytoplasm to induce reprogramming. Second, when we carefully synchronized the cell cycle stage between the transplanted nucleus (ES cell, fetal fibroblast or terminally differentiated cumulus cell) and the recipient I2C cytoplasm, the reconstructed SCNT embryos developed into blastocysts and ES cells capable of contributing to traditional germline and tetraploid chimaeras. Last, direct transfer of cloned embryos, reconstructed with ES cell nuclei, into recipients resulted in live offspring. Thus, the cytoplasm of I2C supports efficient reprogramming, with cell cycle synchronization between the donor nucleus and recipient cytoplasm as the most critical parameter determining success. The ability to use interphase cytoplasm in SCNT could aid efforts to generate autologous human ES cells for regenerative applications, as donated or discarded embryos are more accessible than unfertilized MII oocytes.


Subject(s)
Cellular Reprogramming , Cytoplasm/metabolism , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Interphase , Nuclear Transfer Techniques , Animals , Cell Count , Cloning, Organism , Female , Male , Metaphase , Mice
3.
Fertil Steril ; 101(1): 31-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24382342

ABSTRACT

The exchange of nuclear genetic material between oocytes and embryos offers a novel reproductive option for the prevention of inherited mitochondrial diseases. Mitochondrial dysfunction has been recognized as a significant cause of a number of serious multiorgan diseases. Tissues with a high metabolic demand, such as brain, heart, muscle, and central nervous system, are often affected. Mitochondrial disease can be due to mutations in mitochondrial DNA or in nuclear genes involved in mitochondrial function. There is no curative treatment for patients with mitochondrial disease. Given the lack of treatments and the limitations of prenatal and preimplantation diagnosis, attention has focused on prevention of transmission of mitochondrial disease through germline gene replacement therapy. Because mitochondrial DNA is strictly maternally inherited, two approaches have been proposed. In the first, the nuclear genome from the pronuclear stage zygote of an affected woman is transferred to an enucleated donor zygote. A second technique involves transfer of the metaphase II spindle from the unfertilized oocyte of an affected woman to an enucleated donor oocyte. Our group recently reported successful spindle transfer between human oocytes, resulting in blastocyst development and embryonic stem cell derivation, with very low levels of heteroplasmy. In this review we summarize these novel assisted reproductive techniques and their use to prevent transmission of mitochondrial disorders. The promises and challenges are discussed, focusing on their potential clinical application.


Subject(s)
Fertilization in Vitro/methods , Genetic Therapy/methods , Mitochondrial Diseases/genetics , Mitochondrial Diseases/prevention & control , Animals , Female , Fertilization in Vitro/trends , Genetic Therapy/trends , Humans , Male , Nuclear Transfer Techniques/trends , Reproductive Techniques, Assisted/trends
4.
Cell ; 153(6): 1228-38, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23683578

ABSTRACT

Reprogramming somatic cells into pluripotent embryonic stem cells (ESCs) by somatic cell nuclear transfer (SCNT) has been envisioned as an approach for generating patient-matched nuclear transfer (NT)-ESCs for studies of disease mechanisms and for developing specific therapies. Past attempts to produce human NT-ESCs have failed secondary to early embryonic arrest of SCNT embryos. Here, we identified premature exit from meiosis in human oocytes and suboptimal activation as key factors that are responsible for these outcomes. Optimized SCNT approaches designed to circumvent these limitations allowed derivation of human NT-ESCs. When applied to premium quality human oocytes, NT-ESC lines were derived from as few as two oocytes. NT-ESCs displayed normal diploid karyotypes and inherited their nuclear genome exclusively from parental somatic cells. Gene expression and differentiation profiles in human NT-ESCs were similar to embryo-derived ESCs, suggesting efficient reprogramming of somatic cells to a pluripotent state.


Subject(s)
Cell Line , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Nuclear Transfer Techniques , Adult , Animals , Blastocyst/cytology , Cell Fusion , Cell Nucleus/genetics , Cell Separation , Female , Fetus/cytology , Humans , Macaca mulatta , Mitochondria/genetics , Oocytes/cytology , Oocytes/metabolism , Skin/cytology
5.
Nature ; 493(7434): 627-31, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23103867

ABSTRACT

Mutations in mitochondrial DNA (mtDNA) are associated with severe human diseases and are maternally inherited through the egg's cytoplasm. Here we investigated the feasibility of mtDNA replacement in human oocytes by spindle transfer (ST; also called spindle-chromosomal complex transfer). Of 106 human oocytes donated for research, 65 were subjected to reciprocal ST and 33 served as controls. Fertilization rate in ST oocytes (73%) was similar to controls (75%); however, a significant portion of ST zygotes (52%) showed abnormal fertilization as determined by an irregular number of pronuclei. Among normally fertilized ST zygotes, blastocyst development (62%) and embryonic stem cell isolation (38%) rates were comparable to controls. All embryonic stem cell lines derived from ST zygotes had normal euploid karyotypes and contained exclusively donor mtDNA. The mtDNA can be efficiently replaced in human oocytes. Although some ST oocytes displayed abnormal fertilization, remaining embryos were capable of developing to blastocysts and producing embryonic stem cells similar to controls.


Subject(s)
Genetic Therapy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/therapy , Nuclear Transfer Techniques/standards , Adult , Animals , Cell Nucleus/genetics , Cryopreservation , Cytoplasm/genetics , DNA, Mitochondrial/analysis , DNA, Mitochondrial/genetics , Embryo, Mammalian/embryology , Embryonic Stem Cells/cytology , Female , Fertilization , Humans , Macaca mulatta/genetics , Macaca mulatta/growth & development , Microsatellite Repeats/genetics , Oocytes/cytology , Pregnancy , Young Adult , Zygote/cytology , Zygote/pathology
6.
Dev Biol ; 371(2): 146-55, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22935618

ABSTRACT

Inactivation of one X chromosome in female mammals (XX) compensates for the reduced dosage of X-linked gene expression in males (XY). However, the inner cell mass (ICM) of mouse preimplantation blastocysts and their in vitro counterparts, pluripotent embryonic stem cells (ESCs), initially maintain two active X chromosomes (XaXa). Random X chromosome inactivation (XCI) takes place in the ICM lineage after implantation or upon differentiation of ESCs, resulting in mosaic tissues composed of two cell types carrying either maternal or paternal active X chromosomes. While the status of XCI in human embryos and ICMs remains unknown, majority of human female ESCs show non-random XCI. We demonstrate here that rhesus monkey ESCs also display monoallelic expression and methylation of X-linked genes in agreement with non-random XCI. However, XIST and other X-linked genes were expressed from both chromosomes in isolated female monkey ICMs indicating that ex vivo pluripotent cells retain XaXa. Intriguingly, the trophectoderm (TE) in preimplantation monkey blastocysts also expressed X-linked genes from both alleles suggesting that, unlike the mouse, primate TE lineage does not support imprinted paternal XCI. Our results provide insights into the species-specific nature of XCI in the primate system and reveal fundamental epigenetic differences between in vitro and ex vivo primate pluripotent cells.


Subject(s)
Embryo, Mammalian/metabolism , Pluripotent Stem Cells/metabolism , X Chromosome Inactivation , X Chromosome/genetics , Animals , Blastocyst/metabolism , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Genes, X-Linked , Genomic Imprinting , Macaca mulatta , Male
7.
Cell Rep ; 1(5): 506-15, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22701816

ABSTRACT

The timing and mechanisms of mitochondrial DNA (mtDNA) segregation and transmission in mammals are poorly understood. Genetic bottleneck in female germ cells has been proposed as the main phenomenon responsible for rapid intergenerational segregation of heteroplasmic mtDNA. We demonstrate here that mtDNA segregation occurs during primate preimplantation embryogenesis resulting in partitioning of mtDNA variants between daughter blastomeres. A substantial shift toward homoplasmy occurred in fetuses and embryonic stem cells (ESCs) derived from these heteroplasmic embryos. We also observed a wide range of heteroplasmic mtDNA variants distributed in individual oocytes recovered from these fetuses. Thus, we present here evidence for a previously unknown mtDNA segregation and bottleneck during preimplantation embryo development, suggesting that return to the homoplasmic condition can occur during development of an individual organism from the zygote to birth, without a passage through the germline.


Subject(s)
Blastocyst/cytology , Cell Division/genetics , DNA, Mitochondrial/genetics , Embryonic Development/genetics , Haplotypes/genetics , Macaca mulatta/embryology , Oocytes/cytology , Animals , Blastocyst/metabolism , Blastomeres/cytology , Blastomeres/metabolism , DNA, Mitochondrial/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Gene Dosage/genetics , Macaca mulatta/genetics , Macaca mulatta/metabolism , Oocytes/metabolism , Pregnancy
8.
Fertil Steril ; 97(5): e16, 2012 May.
Article in English | MEDLINE | ID: mdl-22542144

ABSTRACT

OBJECTIVE: To demonstrate step-by-step micromanipulation procedures required for transfer of spindle-chromosomal complexes between mature oocytes. DESIGN: Video presentation of reproductive biology study. SETTING: In vitro fertilization and embryo manipulation laboratory. ANIMAL(S): Rhesus (Macaca mulatta) primates. INTERVENTION(S): Transplantation of the genetic material between mammalian oocytes offers many opportunities to study various aspects of nuclear-cytoplasmic interactions during oogenesis, fertilization and embryo development. We demonstrate the feasibility of isolation and transfer of chromosomes between mature metaphase II (MII) primate oocyte. After fertilization, manipulated oocytes were capable of producing healthy offspring or embryonic stem cells. MAIN OUTCOME MEASURE(S): In this video, we show micromanipulation procedures required for isolation and transfer of spindle-chromosomal complexes between rhesus MII oocytes. In brief, the spindle is visualized using a polarized microscope and extracted into a membrane enclosed karyoplast. Karyoplasts are then reintroduced into an enucleated recipient oocyte (cytoplast, derived from an another female) by karyoplast-cytoplast membrane fusion. RESULT(S): Newly reconstructed oocytes consist of nuclear genetic material from one female and cytoplasmic components, including mitochondria and mitochondrial DNA from another. CONCLUSION(S): This video demonstrates the protocol developed for primate oocytes that successfully allowed of isolation and transfer of chromosomes between mature metaphase II (MII) oocytes. Potential clinical applications include mitochondrial gene replacement therapy to prevent transmission of mtDNA mutations and treatment of infertility caused by cytoplasmic defects in oocytes. Video is available at http://fertstertforum.com/2012974tachibana/.


Subject(s)
Chromosomes/ultrastructure , Gene Transfer Techniques , Micromanipulation , Oocytes/ultrastructure , Spindle Apparatus/transplantation , Animals , Feasibility Studies , Female , Macaca mulatta , Membrane Fusion , Metaphase , Microscopy, Polarization , Spindle Apparatus/ultrastructure
9.
Cell ; 148(1-2): 285-95, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22225614

ABSTRACT

Totipotent cells in early embryos are progenitors of all stem cells and are capable of developing into a whole organism, including extraembryonic tissues such as placenta. Pluripotent cells in the inner cell mass (ICM) are the descendants of totipotent cells and can differentiate into any cell type of a body except extraembryonic tissues. The ability to contribute to chimeric animals upon reintroduction into host embryos is the key feature of murine totipotent and pluripotent cells. Here, we demonstrate that rhesus monkey embryonic stem cells (ESCs) and isolated ICMs fail to incorporate into host embryos and develop into chimeras. However, chimeric offspring were produced following aggregation of totipotent cells of the four-cell embryos. These results provide insights into the species-specific nature of primate embryos and suggest that a chimera assay using pluripotent cells may not be feasible.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Chimera , Embryonic Stem Cells/cytology , Macaca mulatta , Animals , Embryo, Mammalian/cytology , Species Specificity
10.
Nat Protoc ; 5(6): 1138-47, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20539289

ABSTRACT

In this article, we describe detailed protocols for the isolation and transfer of spindle-chromosomal complexes between mature, metaphase II-arrested oocytes. In brief, the spindle-chromosomal complex is visualized using a polarized microscope and extracted into a membrane-enclosed karyoplast. Chromosomes are then reintroduced into an enucleated recipient egg (cytoplast), derived from another female, by karyoplast-cytoplast membrane fusion. Newly reconstructed oocytes consist of nuclear genetic material from one female and cytoplasmic components, including mitochondria and mitochondrial DNA (mtDNA), from another female. This approach yields developmentally competent oocytes suitable for fertilization and producing embryonic stem cells or healthy offspring. The protocol was initially developed for monkey oocytes but can also be used in other species, including mouse and human oocytes. Potential clinical applications include mitochondrial gene replacement therapy to prevent transmission of mtDNA mutations and treatment of infertility caused by cytoplasmic defects in oocytes. Chromosome transfer between the cohorts of oocytes isolated from two females can be completed within 2 h.


Subject(s)
Chromosomes/ultrastructure , Nuclear Transfer Techniques , Oocytes/ultrastructure , Animals , Chromosomes/genetics , Female , Gene Transfer Techniques , Humans , In Vitro Techniques , Macaca mulatta , Male , Metaphase/genetics , Mice , Micromanipulation/instrumentation , Micromanipulation/methods , Nuclear Transfer Techniques/instrumentation , Oocytes/cytology , Sperm Injections, Intracytoplasmic
11.
Int J Dev Biol ; 54(11-12): 1671-8, 2010.
Article in English | MEDLINE | ID: mdl-21404187

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.


Subject(s)
Cloning, Organism/methods , Nuclear Transfer Techniques , Primates/embryology , Animals , Blastocyst , Cellular Reprogramming , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
12.
Dev Biol ; 335(1): 179-87, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19733166

ABSTRACT

The first lineage decision during mammalian development is the establishment of the trophectoderm (TE) and the inner cell mass (ICM). The caudal-type homeodomain protein Cdx2 is implicated in the formation and maintenance of the TE in the mouse. However, the role of CDX2 during early embryonic development in primates is unknown. Here, we demonstrated that CDX2 mRNA levels were detectable in rhesus monkey oocytes, significantly upregulated in pronuclear stage zygotes, diminished in early cleaving embryos but restored again in compact morula and blastocyst stages. CDX2 protein was localized to the nucleus of TE cells but absent altogether in the ICM. Knockdown of CDX2 in monkey oocytes resulted in formation of early blastocyst-like embryos that failed to expand and ceased development. However, the ICM lineage of CDX2-deficient embryos supported the isolation of functional embryonic stem cells. These results provide evidence that CDX2 plays an essential role in functional TE formation during primate embryonic development.


Subject(s)
Cell Lineage , Embryo, Mammalian , Macaca mulatta , Morphogenesis/physiology , Animals , Biomarkers/metabolism , CDX2 Transcription Factor , Cell Differentiation/physiology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Macaca mulatta/anatomy & histology , Macaca mulatta/embryology , Mice , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Oocytes/cytology , Oocytes/physiology
13.
Nature ; 461(7262): 367-72, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19710649

ABSTRACT

Mitochondria are found in all eukaryotic cells and contain their own genome (mitochondrial DNA or mtDNA). Unlike the nuclear genome, which is derived from both the egg and sperm at fertilization, the mtDNA in the embryo is derived almost exclusively from the egg; that is, it is of maternal origin. Mutations in mtDNA contribute to a diverse range of currently incurable human diseases and disorders. To establish preclinical models for new therapeutic approaches, we demonstrate here that the mitochondrial genome can be efficiently replaced in mature non-human primate oocytes (Macaca mulatta) by spindle-chromosomal complex transfer from one egg to an enucleated, mitochondrial-replete egg. The reconstructed oocytes with the mitochondrial replacement were capable of supporting normal fertilization, embryo development and produced healthy offspring. Genetic analysis confirmed that nuclear DNA in the three infants born so far originated from the spindle donors whereas mtDNA came from the cytoplast donors. No contribution of spindle donor mtDNA was detected in offspring. Spindle replacement is shown here as an efficient protocol replacing the full complement of mitochondria in newly generated embryonic stem cell lines. This approach may offer a reproductive option to prevent mtDNA disease transmission in affected families.


Subject(s)
DNA, Mitochondrial/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Mitochondrial/genetics , Genome, Mitochondrial/genetics , Macaca mulatta/genetics , Reproductive Techniques, Assisted , Animals , Cell Nucleus/genetics , DNA, Mitochondrial/analysis , Embryo Transfer , Embryonic Stem Cells/transplantation , Female , Fertilization in Vitro , Macaca mulatta/embryology , Male , Meiosis , Mitochondrial Diseases/genetics , Mitochondrial Diseases/prevention & control , Mutation , Oocytes/cytology , Oocytes/metabolism , Pregnancy
14.
Stem Cells ; 27(6): 1255-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19489081

ABSTRACT

We recently demonstrated that somatic cells from adult primates could be reprogrammed into a pluripotent state by somatic cell nuclear transfer. However, the low efficiency with donor cells from one monkey necessitated the need for large oocyte numbers. Here, we demonstrate nearly threefold higher blastocyst development and embryonic stem (ES) cell derivation rates with different nuclear donor cells. Two ES cell lines were isolated using adult female rhesus macaque skin fibroblasts as nuclear donors and oocytes retrieved from one female, following a single controlled ovarian stimulation. In addition to routine pluripotency tests involving in vitro and in vivo differentiation into various somatic cell types, primate ES cells derived from reprogrammed somatic cells were also capable of contributing to cells expressing markers of germ cells. Moreover, imprinted gene expression, methylation, telomere length, and X-inactivation analyses were consistent with accurate and extensive epigenetic reprogramming of somatic cells by oocyte-specific factors.


Subject(s)
Epigenesis, Genetic , Fibroblasts/cytology , Nuclear Transfer Techniques , Oocytes/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Female , Gene Expression , Macaca mulatta , Reverse Transcriptase Polymerase Chain Reaction
15.
Am J Primatol ; 71(5): 384-92, 2009 May.
Article in English | MEDLINE | ID: mdl-19189308

ABSTRACT

To date, ultrasonography of monkey ovaries is rare and typically of low resolution. The objectives of this study were to use state-of-the-art, high-resolution, transabdominal ultrasonography with real-time Doppler capabilities to: (1) determine whether one can reliably detect in real time the large dominant follicle, the corpus luteum (CL), and small (<2 mm) antral follicles on the ovaries of rhesus monkeys during the natural menstrual cycle; and (2) predict the follicular response of rhesus ovaries to controlled ovarian stimulation (COS) protocols. Rhesus monkeys were selected for transabdominal ultrasonography using a GE Voluson 730 Expert Doppler System at discrete stages of the menstrual cycle. Subsequently, serial ultrasound scanning was employed to observe growth of antral follicles and the CL. Finally, females were scanned to assess follicular growth during COS. The dominant structure and small antral follicles (<2 mm) were reliably visualized in real time. The follicle destined to ovulate could be identified by size differential by day 3 of the follicular phase. The number of small antral follicles present before onset of COS protocol correlated positively with the number of metaphase II-stage oocytes collected after treatment. The results of this study demonstrate that the population dynamics of antral follicle pools can be noninvasively evaluated in monkeys during natural and pharmacologic ovarian cycles.


Subject(s)
Estrous Cycle/physiology , Macaca mulatta/physiology , Ovarian Follicle/diagnostic imaging , Ovulation Induction/veterinary , Ultrasonography, Doppler/veterinary , Animals , Estradiol/blood , Female , Longitudinal Studies , Ovarian Follicle/physiology , Progesterone/blood
16.
Am J Primatol ; 69(8): 917-29, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17358011

ABSTRACT

The vervet monkey was evaluated as a primate model for use in assisted reproductive technologies (ARTs). Eight adult female vervets were hormonally monitored for their potential use as egg donors and those six females displaying regular menstrual cycles were subjected to controlled ovarian stimulation with recombinant human gonadotropins. Three animals failed to respond while laparoscopic follicular aspiration was performed on the other three females at 27-30 h post-human chorionic gonadotropin administration. A total of 62, 40, and 18 oocytes was recovered from these three animals of which 30, 20, and 4, respectively, matured to the metaphase II stage and were subsequently inseminated using intracytoplasmic sperm injection. An average of 40+/-15% (SEM) of the inseminated oocytes were fertilized based on pronucleus formation and timely cleavage. One embryo from each of the two stimulated females developed into expanded blastocysts. Two adult male vervets were assessed as sperm donors. Neither adjusted well to the restraint and collection procedure required for penile electroejaculation. Samples collected via rectal electroejaculation were very low in sperm motility and concentration; however, cauda epididymal aspirations from one male yielded an adequate concentration of motile sperm. These results emphasize the need to establish species-specific ovarian stimulation protocols and semen collection techniques if vervets are to be considered for basic and applied (ARTs) research on primate gametes or embryos.


Subject(s)
Cercopithecinae , Models, Animal , Sperm Injections, Intracytoplasmic , Animals , Blastocyst/cytology , Ejaculation , Embryo Transfer , Embryonic Development , Female , Male , Ovarian Follicle/diagnostic imaging , Ovulation Induction , Species Specificity , Sperm Motility , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...