Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
Hum Reprod ; 35(4): 743-750, 2020 04 28.
Article En | MEDLINE | ID: mdl-32296829

STUDY QUESTION: What proportion of fertilized human ova are lost before implantation? SUMMARY ANSWER: An estimated 40 to 50% of fertilized ova fail to implant. WHAT IS KNOWN ALREADY: Preimplantation loss is not detectable with current technology. Published estimates of preimplantation loss range from 10 to 70%. STUDY DESIGN, SIZE, DURATION: We combine data from epidemiologic, demographic, laboratory and in vitro fertilization studies to construct an empirical framework for the estimation of preimplantation loss. This framework is summarized in a user-friendly Excel file included in supplement. PARTICIPANTS/MATERIALS, SETTING, METHODS: We draw from multiple sources to generate plausible estimates of fecundability, sterility, transient anovulation, intercourse patterns and the proportion of ova fertilized in the presence of sperm. We combine these estimates to generate a summary estimate of preimplantation loss. This estimate can be considered an average for couples in their prime reproductive years. MAIN RESULTS AND THE ROLE OF CHANCE: Under a plausible range of assumptions, we estimate that 40 to 50% of fertilized ova fail to implant. LIMITATIONS, REASONS FOR CAUTION: A crucial factor in estimating preimplantation loss is the probability that an ovum will be fertilized when exposed to sperm. Human data are available only from in vitro fertilization (IVF), which may not accurately represent events in vivo. We therefore assume a range of in vivo fertilization rates, from 64% (human IVF data) to 90% (mouse data). WIDER IMPLICATIONS OF THE FINDINGS: Our estimate of preimplantation loss takes into account the biological processes relevant to fertilization and loss. Using this empirical basis for estimation, we find support for the usual assumption that risk of loss is highest in the earliest days following fertilization. Furthermore, this framework can provide improved estimates as better reproductive data become available. To the extent that our estimates are accurate, more fertilized ova are apparently lost in vitro than in vivo, suggesting that further improvements in IVF success rates may be possible. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the Intramural Program of the National Institute of Environmental Health Sciences, NIH. Professor Adashi serves as Co-Chair of the Safety Advisory Board of Ohana Biosciences, Inc. The other authors have no competing interests. TRIAL REGISTRATION NUMBER: N/A.


Embryo Implantation , Fertilization in Vitro , Animals , Female , Fertilization , Humans , Male , Mice , Ovalbumin , Spermatozoa
2.
Nat Med ; 25(6): 890-897, 2019 06.
Article En | MEDLINE | ID: mdl-31160821

Monogenic disorders occur at a high frequency in human populations and are commonly inherited through the germline. Unfortunately, once the mutation has been transmitted to a child, only limited treatment options are available in most cases. However, means of correcting disease-causing nuclear and mitochondrial DNA mutations in gametes or preimplantation embryos have now been developed and are commonly referred to as germline gene therapy (GGT). We will discuss these novel strategies and provide a path forward for safe, high-efficiency GGT that may provide a promising new paradigm for preventing the passage of deleterious genes from parent to child.


Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Germ-Line Mutation , Child , DNA Repair , DNA, Mitochondrial/genetics , Female , Fertilization in Vitro , Gene Conversion , Genetic Therapy/ethics , Genetic Therapy/legislation & jurisprudence , Humans , Male , Mitochondrial Replacement Therapy , Pregnancy , Preimplantation Diagnosis , Safety
5.
PLoS One ; 13(7): e0201304, 2018.
Article En | MEDLINE | ID: mdl-30040856

The accumulation of acquired mitochondrial genome (mtDNA) mutations with aging in somatic cells has been implicated in mitochondrial dysfunction and linked to age-onset diseases in humans. Here, we asked if somatic mtDNA mutations are also associated with aging in the mouse. MtDNA integrity in multiple organs and tissues in young and old (2-34 months) wild type (wt) mice was investigated by whole genome sequencing. Remarkably, no acquired somatic mutations were detected in tested tissues. However, we identified several non-synonymous germline mtDNA variants whose heteroplasmy levels (ratio of normal to mutant mtDNA) increased significantly with aging suggesting clonal expansion of inherited mtDNA mutations. Polg mutator mice, a model for premature aging, exhibited both germline and somatic mtDNA mutations whose numbers and heteroplasmy levels increased significantly with age implicating involvement in premature aging. Our results suggest that, in contrast to humans, acquired somatic mtDNA mutations do not accompany the aging process in wt mice.


Aging , DNA, Mitochondrial/genetics , Mice/genetics , Mutation , Aging, Premature/genetics , Aging, Premature/veterinary , Animals , DNA Polymerase gamma/genetics , Female , Germ Cells/metabolism , Male , Mice/embryology , Mice/physiology , Mice, Inbred C57BL , Mitochondria/genetics
7.
J Assist Reprod Genet ; 34(11): 1427-1434, 2017 Nov.
Article En | MEDLINE | ID: mdl-28942525

PURPOSE: The main purposes of the study were to investigate the endocrine function of ovarian tissue transplanted to heterotopic subcutaneous sites and the reproductive competence and telomere length of a nonhuman primate originating from transplanted tissue. METHODS: Ovarian cortex pieces were transplanted into the original rhesus macaques in the arm subcutaneously, in the abdomen next to muscles, or in the kidney. Serum estradiol (E2) and progesterone (P4) concentrations were measured weekly for up to 8 years following tissue transplantation. A monkey derived from an oocyte in transplanted ovarian tissue entered time-mated breeding and underwent controlled ovarian stimulation. Pregnancy and offspring were evaluated. Telomere lengths and oocytes obtained following controlled ovarian stimulation were assessed. RESULTS: Monkeys with transplants in the arm and abdomen had cyclic E2 of 100 pg/ml, while an animal with arm transplants had E2 of 50 pg/ml. One monkey with transplants in the abdomen and kidney had ovulatory cycles for 3 years. A monkey derived from an oocyte in transplanted tissue conceived and had a normal gestation until intrapartum fetal demise. She conceived again and delivered a healthy offspring at term. Controlled ovarian stimulations of this monkey yielded mature oocytes comparable to controls. Her telomere length was long relative to controls. CONCLUSIONS: Heterotopic ovarian tissue transplants yielded long-term endocrine function in macaques. A monkey derived from an oocyte in transplanted tissue was reproductively competent. Her telomere length did not show epigenetically induced premature cellular aging. Ovarian tissue transplantation to heterotopic sites for fertility preservation should move forward cautiously, yet optimistically.


Fertility Preservation/methods , Oocytes/growth & development , Ovarian Follicle/transplantation , Ovary/transplantation , Reproduction/physiology , Animals , Cryopreservation , Estradiol/blood , Female , Macaca mulatta/genetics , Macaca mulatta/physiology , Ovarian Follicle/growth & development , Ovary/growth & development , Ovulation Induction/methods , Pregnancy , Progesterone/blood , Reproduction/genetics , Telomere Homeostasis/genetics
8.
Nature ; 548(7668): 413-419, 2017 08 24.
Article En | MEDLINE | ID: mdl-28783728

Genome editing has potential for the targeted correction of germline mutations. Here we describe the correction of the heterozygous MYBPC3 mutation in human preimplantation embryos with precise CRISPR-Cas9-based targeting accuracy and high homology-directed repair efficiency by activating an endogenous, germline-specific DNA repair response. Induced double-strand breaks (DSBs) at the mutant paternal allele were predominantly repaired using the homologous wild-type maternal gene instead of a synthetic DNA template. By modulating the cell cycle stage at which the DSB was induced, we were able to avoid mosaicism in cleaving embryos and achieve a high yield of homozygous embryos carrying the wild-type MYBPC3 gene without evidence of off-target mutations. The efficiency, accuracy and safety of the approach presented suggest that it has potential to be used for the correction of heritable mutations in human embryos by complementing preimplantation genetic diagnosis. However, much remains to be considered before clinical applications, including the reproducibility of the technique with other heterozygous mutations.


Carrier Proteins/genetics , Embryo, Mammalian/metabolism , Gene Editing/methods , Mutation/genetics , Adult , Alleles , Blastocyst/metabolism , Blastocyst/pathology , Cell Division , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Breaks, Double-Stranded , Embryo, Mammalian/pathology , Gene Targeting , Genetic Complementation Test , Heterozygote , Homozygote , Humans , Male , Mosaicism , Recombinational DNA Repair/genetics , S Phase , Templates, Genetic , Zygote/metabolism , Zygote/pathology
9.
EMBO J ; 36(15): 2177-2181, 2017 08 01.
Article En | MEDLINE | ID: mdl-28679504

Mitochondria, the ubiquitous power packs in nearly every eukaryotic cell, contain their own DNA, known as mtDNA, which is inherited exclusively from the mother. The number of mitochondrial genomes varies depending on the cell's energy needs. The mature oocyte contains the highest number of mitochondria of any cell type, although there is little if any mtDNA replication after fertilization until the embryo implants. This has potential repercussions for mitochondrial replacement therapy (MRT; see description of currently employed methods below) used to prevent the transmission of mtDNA-based disorders. If only a few mitochondria with defective mtDNA are left in the embryo and undergo extensive replication, it might therefore thwart the purpose of MRT In order to improve the safety and efficacy of this experimental therapy, we need a better understanding of how and which mtDNA is tagged for replication versus transcription after fertilization of the oocyte.


DNA Replication , Genome, Human , Genome, Mitochondrial , Germ Cells , Wills , Humans , Mitochondrial Replacement Therapy/methods , Models, Biological
10.
Stem Cells ; 35(1): 26-34, 2017 01.
Article En | MEDLINE | ID: mdl-27612640

Embryonic stem cells (ESC) hold promise for the treatment of human medical conditions but are allogeneic. Here, we consider the differences between autologous pluripotent stem cells produced by nuclear transfer (NT-ESCs) and transcription factor-mediated, induced pluripotent stem cells (iPSCs) that impact the desirability of each of these cell types for clinical use. The derivation of NT-ESCs is more cumbersome and requires donor oocytes; however, the use of oocyte cytoplasm as the source of reprogramming factors is linked to a key advantage of NT-ESCs-the ability to replace mutant mitochondrial DNA in a patient cell (due to either age or inherited disease) with healthy donor mitochondria from an oocyte. Moreover, in epigenomic and transcriptomic comparisons between isogenic iPSCs and NT-ESCs, the latter produced cells that more closely resemble bona fide ESCs derived from fertilized embryos. Thus, although NT-ESCs are more difficult to generate than iPSCs, the ability of somatic cell nuclear transfer to replace aged or diseased mitochondria and the closer epigenomic and transcriptomic similarity between NT-ESCs and bona fide ESCs may make NT-ESCs superior for future applications in regenerative medicine. Stem Cells 2017;35:26-34.


Embryonic Stem Cells/cytology , Nuclear Transfer Techniques , Animals , Clinical Trials as Topic , DNA, Mitochondrial/genetics , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism
11.
Cell Stem Cell ; 20(1): 112-119, 2017 01 05.
Article En | MEDLINE | ID: mdl-27840020

Oocyte defects lie at the heart of some forms of infertility and could potentially be addressed therapeutically by alternative routes for oocyte formation. Here, we describe the generation of functional human oocytes following nuclear transfer of first polar body (PB1) genomes from metaphase II (MII) oocytes into enucleated donor MII cytoplasm (PBNT). The reconstructed oocytes supported the formation of de novo meiotic spindles and, after fertilization with sperm, meiosis completion and formation of normal diploid zygotes. While PBNT zygotes developed to blastocysts less frequently (42%) than controls (75%), genome-wide genetic, epigenetic, and transcriptional analyses of PBNT and control ESCs indicated comparable numbers of structural variations and markedly similar DNA methylation and transcriptome profiles. We conclude that rescue of PB1 genetic material via introduction into donor cytoplasm may offer a source of oocytes for infertility treatment or mitochondrial replacement therapy for mtDNA disease.


Genome, Human , Nuclear Transfer Techniques , Oocytes/metabolism , Polar Bodies/metabolism , Adult , Blastocyst/metabolism , DNA Methylation/genetics , Embryonic Development/genetics , Epigenesis, Genetic , Female , Fertilization in Vitro , Gene Expression Profiling , Genomic Instability , Human Embryonic Stem Cells/metabolism , Humans , Male , Metaphase , Ploidies , Sequence Analysis, RNA , Spermatozoa/metabolism , Spindle Apparatus/metabolism , Transcription, Genetic
12.
Nature ; 540(7632): 270-275, 2016 12 08.
Article En | MEDLINE | ID: mdl-27919073

Maternally inherited mitochondrial (mt)DNA mutations can cause fatal or severely debilitating syndromes in children, with disease severity dependent on the specific gene mutation and the ratio of mutant to wild-type mtDNA (heteroplasmy) in each cell and tissue. Pathogenic mtDNA mutations are relatively common, with an estimated 778 affected children born each year in the United States. Mitochondrial replacement therapies or techniques (MRT) circumventing mother-to-child mtDNA disease transmission involve replacement of oocyte maternal mtDNA. Here we report MRT outcomes in several families with common mtDNA syndromes. The mother's oocytes were of normal quality and mutation levels correlated with those in existing children. Efficient replacement of oocyte mutant mtDNA was performed by spindle transfer, resulting in embryos containing >99% donor mtDNA. Donor mtDNA was stably maintained in embryonic stem cells (ES cells) derived from most embryos. However, some ES cell lines demonstrated gradual loss of donor mtDNA and reversal to the maternal haplotype. In evaluating donor-to-maternal mtDNA interactions, it seems that compatibility relates to mtDNA replication efficiency rather than to mismatch or oxidative phosphorylation dysfunction. We identify a polymorphism within the conserved sequence box II region of the D-loop as a plausible cause of preferential replication of specific mtDNA haplotypes. In addition, some haplotypes confer proliferative and growth advantages to cells. Hence, we propose a matching paradigm for selecting compatible donor mtDNA for MRT.


DNA, Mitochondrial/genetics , DNA, Mitochondrial/therapeutic use , Maternal Inheritance/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Replacement Therapy/methods , Mutation , Oocytes/metabolism , Blastocyst/cytology , Blastocyst/metabolism , Cell Line , Conserved Sequence/genetics , DNA, Mitochondrial/biosynthesis , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Haplotypes/genetics , Humans , Male , Meiosis , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/prevention & control , Oocyte Donation , Oocytes/cytology , Oocytes/pathology , Oxidative Phosphorylation , Pedigree , Polymorphism, Genetic
13.
Cell Metab ; 24(2): 283-94, 2016 08 09.
Article En | MEDLINE | ID: mdl-27425585

Vertebrate cells carry two different genomes, nuclear (nDNA) and mitochondrial (mtDNA), both encoding proteins involved in oxidative phosphorylation. Because of the extensive interactions, adaptive coevolution of the two genomes must occur to ensure normal mitochondrial function. To investigate whether incompatibilities between these two genomes could contribute to interspecies reproductive barriers, we performed reciprocal mtDNA replacement (MR) in zygotes between widely divergent Mus m. domesticus (B6) and conplastic Mus m. musculus (PWD) mice. Transfer of MR1 cybrid embryos (B6nDNA-PWDmtDNA) supported normal development of F1 offspring with reduced male fertility but unaffected reproductive fitness in females. Furthermore, donor PWD mtDNA was faithfully transmitted through the germline into F2 and F3 generations. In contrast, reciprocal MR2 (PWDnDNA-B6mtDNA) produced high embryonic loss and stillborn rates, suggesting an association between mitochondrial function and infertility. These results strongly suggest that functional incompatibility between nuclear and mitochondrial genomes contributes to interspecies reproductive isolation in mammals.


Cell Nucleus/genetics , Genome, Mitochondrial , Reproduction/genetics , Animals , Blastocyst/metabolism , Crosses, Genetic , DNA, Mitochondrial/genetics , Embryo Loss/genetics , Embryonic Development/genetics , Female , Germ Cells/metabolism , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Species Specificity
14.
Cell Stem Cell ; 18(5): 625-36, 2016 05 05.
Article En | MEDLINE | ID: mdl-27151456

The genetic integrity of iPSCs is an important consideration for therapeutic application. In this study, we examine the accumulation of somatic mitochondrial genome (mtDNA) mutations in skin fibroblasts, blood, and iPSCs derived from young and elderly subjects (24-72 years). We found that pooled skin and blood mtDNA contained low heteroplasmic point mutations, but a panel of ten individual iPSC lines from each tissue or clonally expanded fibroblasts carried an elevated load of heteroplasmic or homoplasmic mutations, suggesting that somatic mutations randomly arise within individual cells but are not detectable in whole tissues. The frequency of mtDNA defects in iPSCs increased with age, and many mutations were non-synonymous or resided in RNA coding genes and thus can lead to respiratory defects. Our results highlight a need to monitor mtDNA mutations in iPSCs, especially those generated from older patients, and to examine the metabolic status of iPSCs destined for clinical applications.


Aging/genetics , DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Adult , Aged , Blood Cells/metabolism , Fibroblasts/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Skin/cytology
15.
Nature ; 524(7564): 234-8, 2015 Aug 13.
Article En | MEDLINE | ID: mdl-26176921

Mitochondria have a major role in energy production via oxidative phosphorylation, which is dependent on the expression of critical genes encoded by mitochondrial (mt)DNA. Mutations in mtDNA can cause fatal or severely debilitating disorders with limited treatment options. Clinical manifestations vary based on mutation type and heteroplasmy (that is, the relative levels of mutant and wild-type mtDNA within each cell). Here we generated genetically corrected pluripotent stem cells (PSCs) from patients with mtDNA disease. Multiple induced pluripotent stem (iPS) cell lines were derived from patients with common heteroplasmic mutations including 3243A>G, causing mitochondrial encephalomyopathy and stroke-like episodes (MELAS), and 8993T>G and 13513G>A, implicated in Leigh syndrome. Isogenic MELAS and Leigh syndrome iPS cell lines were generated containing exclusively wild-type or mutant mtDNA through spontaneous segregation of heteroplasmic mtDNA in proliferating fibroblasts. Furthermore, somatic cell nuclear transfer (SCNT) enabled replacement of mutant mtDNA from homoplasmic 8993T>G fibroblasts to generate corrected Leigh-NT1 PSCs. Although Leigh-NT1 PSCs contained donor oocyte wild-type mtDNA (human haplotype D4a) that differed from Leigh syndrome patient haplotype (F1a) at a total of 47 nucleotide sites, Leigh-NT1 cells displayed transcriptomic profiles similar to those in embryo-derived PSCs carrying wild-type mtDNA, indicative of normal nuclear-to-mitochondrial interactions. Moreover, genetically rescued patient PSCs displayed normal metabolic function compared to impaired oxygen consumption and ATP production observed in mutant cells. We conclude that both reprogramming approaches offer complementary strategies for derivation of PSCs containing exclusively wild-type mtDNA, through spontaneous segregation of heteroplasmic mtDNA in individual iPS cell lines or mitochondrial replacement by SCNT in homoplasmic mtDNA-based disease.


DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Haplotypes/genetics , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Mice , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/pathology , Mutation/genetics , Nuclear Transfer Techniques , Nucleotides/genetics , Oxygen Consumption , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, RNA , Skin/cytology
16.
Trends Mol Med ; 21(2): 68-76, 2015 Feb.
Article En | MEDLINE | ID: mdl-25573721

Mitochondrial dysfunction is implicated in disease and age-related infertility. Mitochondrial replacement therapies (MRT) in oocytes or zygotes, such as pronuclear (PNT), spindle (ST), or polar body (PBT) transfer, could prevent second-generation transmission of mitochondrial DNA (mtDNA) defects. PNT, associated with high levels of mtDNA carryover in mice but low levels in human embryos, carries ethical issues secondary to donor embryo destruction. ST, developed in primates, supports normal development to adults and low mtDNA carryover. PBT in mice, coupled with PN or ST, may increase the yield of reconstructed embryos with low mtDNA carryover. MRT also offers replacement of the deficient cytoplasm in oocytes from older patients, with the expectation of high pregnancy rates following in vitro fertilization.


Mitochondria/genetics , Mitochondrial Diseases/therapy , Animals , Biological Therapy , Female , Humans , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/prevention & control , Pregnancy , Reproductive Medicine , Zygote/metabolism
17.
Nature ; 511(7508): 177-83, 2014 Jul 10.
Article En | MEDLINE | ID: mdl-25008523

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.


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
18.
Cell Metab ; 20(1): 6-8, 2014 Jul 01.
Article En | MEDLINE | ID: mdl-24988456

Mitochondrial DNA diseases are relatively common, sometimes devastating, and transmitted exclusively through the egg to children of carrier mothers. A study in Cell by Wang et al. (2014) adds the exciting possibility of a new therapy for preventing mitochondrial disease transmission predicated on the use of polar body genomes in mice.


Mitochondrial Diseases/genetics , Mitochondrial Diseases/prevention & control , Nuclear Transfer Techniques , Oocytes/cytology , Polar Bodies/transplantation , Animals , Humans
19.
Nature ; 509(7498): 101-4, 2014 May 01.
Article En | MEDLINE | ID: mdl-24670652

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.


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
20.
Trends Endocrinol Metab ; 25(1): 5-7, 2014 Jan.
Article En | MEDLINE | ID: mdl-24373414

Inherited diseases caused by mitochondrial gene (mtDNA) mutations affect at least 1 in 5000-10,000 children and are associated with severe clinical symptoms. Novel reproductive techniques designed to replace mutated mtDNA in oocytes or early embryos have been proposed to prevent transmission of disease from parents to their children. Here we review the efficacy and safety of these approaches and their associated ethical and regulatory issues.


DNA, Mitochondrial/genetics , Genes, Mitochondrial , Genetic Therapy/ethics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/prevention & control , Nuclear Transfer Techniques/ethics , Female , Genetic Therapy/legislation & jurisprudence , Humans , Male , Mitochondria/genetics , Mutation , Nuclear Transfer Techniques/legislation & jurisprudence , Oocytes , Reproductive Techniques, Assisted/ethics , Reproductive Techniques, Assisted/legislation & jurisprudence , United Kingdom , United States
...