Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Cell ; 153(6): 1228-38, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23683578

RESUMO

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.


Assuntos
Linhagem Celular , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Técnicas de Transferência Nuclear , Adulto , Animais , Blastocisto/citologia , Fusão Celular , Núcleo Celular/genética , Separação Celular , Feminino , Feto/citologia , Humanos , Macaca mulatta , Mitocôndrias/genética , Oócitos/citologia , Oócitos/metabolismo , Pele/citologia
3.
Nature ; 548(7668): 413-419, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28783728

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Embrião de Mamíferos/metabolismo , Edição de Genes/métodos , Mutação/genética , Adulto , Alelos , Blastocisto/metabolismo , Blastocisto/patologia , Divisão Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Quebras de DNA de Cadeia Dupla , Embrião de Mamíferos/patologia , Marcação de Genes , Teste de Complementação Genética , Heterozigoto , Homozigoto , Humanos , Masculino , Mosaicismo , Reparo de DNA por Recombinação/genética , Fase S , Moldes Genéticos , Zigoto/metabolismo , Zigoto/patologia
4.
Nature ; 540(7632): 270-275, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27919073

RESUMO

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.


Assuntos
DNA Mitocondrial/genética , DNA Mitocondrial/uso terapêutico , Herança Materna/genética , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Terapia de Substituição Mitocondrial/métodos , Mutação , Oócitos/metabolismo , Blastocisto/citologia , Blastocisto/metabolismo , Linhagem Celular , Sequência Conservada/genética , DNA Mitocondrial/biossíntese , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Haplótipos/genética , Humanos , Masculino , Meiose , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/prevenção & controle , Doação de Oócitos , Oócitos/citologia , Oócitos/patologia , Fosforilação Oxidativa , Linhagem , Polimorfismo Genético
5.
EMBO J ; 36(15): 2177-2181, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28679504

RESUMO

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.


Assuntos
Replicação do DNA , Genoma Humano , Genoma Mitocondrial , Células Germinativas , Testamentos , Humanos , Terapia de Substituição Mitocondrial/métodos , Modelos Biológicos
6.
Nature ; 524(7564): 234-8, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26176921

RESUMO

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.


Assuntos
DNA Mitocondrial/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Haplótipos/genética , Humanos , Doença de Leigh/genética , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Camundongos , Mitocôndrias/patologia , Doenças Mitocondriais/patologia , Encefalomiopatias Mitocondriais/genética , Encefalomiopatias Mitocondriais/metabolismo , Encefalomiopatias Mitocondriais/patologia , Mutação/genética , Técnicas de Transferência Nuclear , Nucleotídeos/genética , Consumo de Oxigênio , Polimorfismo de Nucleotídeo Único/genética , Análise de Sequência de RNA , Pele/citologia
7.
Hum Reprod ; 35(4): 743-750, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32296829

RESUMO

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.


Assuntos
Implantação do Embrião , Fertilização in vitro , Animais , Feminino , Fertilização , Humanos , Masculino , Camundongos , Ovalbumina , Espermatozoides
8.
Nature ; 509(7498): 101-4, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24670652

RESUMO

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.


Assuntos
Reprogramação Celular , Citoplasma/metabolismo , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Interfase , Técnicas de Transferência Nuclear , Animais , Contagem de Células , Clonagem de Organismos , Feminino , Masculino , Metáfase , Camundongos
9.
Nature ; 511(7508): 177-83, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25008523

RESUMO

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.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes/metabolismo , Animais , Linhagem Celular , Aberrações Cromossômicas , Cromossomos Humanos X/genética , Cromossomos Humanos X/metabolismo , Variações do Número de Cópias de DNA , Metilação de DNA , Estudo de Associação Genômica Ampla , Impressão Genômica , Humanos , Técnicas de Transferência Nuclear/normas , Células-Tronco Pluripotentes/citologia , Transcriptoma
11.
Stem Cells ; 35(1): 26-34, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27612640

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/citologia , Técnicas de Transferência Nuclear , Animais , Ensaios Clínicos como Assunto , DNA Mitocondrial/genética , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo
12.
J Assist Reprod Genet ; 34(11): 1427-1434, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28942525

RESUMO

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.


Assuntos
Preservação da Fertilidade/métodos , Oócitos/crescimento & desenvolvimento , Folículo Ovariano/transplante , Ovário/transplante , Reprodução/fisiologia , Animais , Criopreservação , Estradiol/sangue , Feminino , Macaca mulatta/genética , Macaca mulatta/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Ovário/crescimento & desenvolvimento , Indução da Ovulação/métodos , Gravidez , Progesterona/sangue , Reprodução/genética , Homeostase do Telômero/genética
14.
Dev Biol ; 335(1): 179-87, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19733166

RESUMO

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.


Assuntos
Linhagem da Célula , Embrião de Mamíferos , Macaca mulatta , Morfogênese/fisiologia , Animais , Biomarcadores/metabolismo , Fator de Transcrição CDX2 , Diferenciação Celular/fisiologia , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Macaca mulatta/anatomia & histologia , Macaca mulatta/embriologia , Camundongos , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Oócitos/citologia , Oócitos/fisiologia
15.
Hum Reprod ; 25(8): 1927-41, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20522441

RESUMO

BACKGROUND: Parthenogenetic embryonic stem cells (PESCs) may have future utilities in cell replacement therapies since they are closely related to the female from which the activated oocyte was obtained. Furthermore, the avoidance of parthenogenetic development in mammals provides the most compelling rationale for the evolution of genomic imprinting, and the biological process of parthenogenesis raises complex issues regarding differential gene expression. METHODS AND RESULTS: We describe here homozygous rhesus monkey PESCs derived from a spontaneously duplicated, haploid oocyte genome. Since the effect of homozygosity on PESCs pluripotency and differentiation potential is unknown, we assessed the similarities and differences in pluripotency markers and developmental potential by in vitro and in vivo differentiation of homozygous and heterozygous PESCs. To understand the differences in gene expression regulation between parthenogenetic and biparental embryonic stem cells (ESCs), we conducted microarray analysis of genome-wide mRNA profiles of primate PESCs and ESCs derived from fertilized embryos using the Affymetrix Rhesus Macaque Genome array. Several known paternally imprinted genes were in the highly down-regulated group in PESCs compared with ESCs. Furthermore, allele-specific expression analysis of other genes whose expression is also down-regulated in PESCs, led to the identification of one novel imprinted gene, inositol polyphosphate-5-phosphatase F (INPP5F), which was exclusively expressed from a paternal allele. CONCLUSION: Our findings suggest that PESCs could be used as a model for studying genomic imprinting, and in the discovery of novel imprinted genes.


Assuntos
Células-Tronco Embrionárias/metabolismo , Impressão Genômica , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/metabolismo , Alelos , Animais , Linhagem Celular , Metilação de DNA , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Triagem de Portadores Genéticos , Humanos , Inositol Polifosfato 5-Fosfatases , Macaca mulatta , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Monoéster Fosfórico Hidrolases/genética
16.
Stem Cells ; 27(6): 1255-64, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489081

RESUMO

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.


Assuntos
Epigênese Genética , Fibroblastos/citologia , Técnicas de Transferência Nuclear , Oócitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Feminino , Expressão Gênica , Macaca mulatta , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Stem Cells ; 26(3): 756-66, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18192229

RESUMO

Monoparental parthenotes represent a potential source of histocompatible stem cells that should be isogenic with the oocyte donor and therefore suitable for use in cell or tissue replacement therapy. We generated five rhesus monkey parthenogenetic embryonic stem cell (PESC) lines with stable, diploid female karyotypes that were morphologically indistinguishable from biparental controls, expressed key pluripotent markers, and generated cell derivatives representative of all three germ layers following in vivo and in vitro differentiation. Interestingly, high levels of heterozygosity were observed at the majority of loci that were polymorphic in the oocyte donors. Some PESC lines were also heterozygous in the major histocompatibility complex region, carrying haplotypes identical to those of the egg donor females. Expression analysis revealed transcripts from some imprinted genes that are normally expressed from only the paternal allele. These results indicate that limitations accompanying the potential use of PESC-derived phenotypes in regenerative medicine, including aberrant genomic imprinting and high levels of homozygosity, are cell line-dependent and not always present. PESC lines were derived in high enough yields to be practicable, and their derivatives are suitable for autologous transplantation into oocyte donors or could be used to establish a bank of histocompatible cell lines for a broad spectrum of patients.


Assuntos
Células-Tronco Embrionárias/citologia , Macaca mulatta/embriologia , Partenogênese , Animais , Diferenciação Celular , Linhagem Celular , Separação Celular , Metilação de DNA , Embrião de Mamíferos , Células-Tronco Embrionárias/metabolismo , Feminino , Fertilização in vitro , Regulação da Expressão Gênica , Impressão Genômica , Genótipo , Heterozigoto , Histocompatibilidade , Humanos , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único/genética , Síndrome de Prader-Willi/genética
18.
Nat Med ; 25(6): 890-897, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31160821

RESUMO

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.


Assuntos
Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Terapia Genética/métodos , Mutação em Linhagem Germinativa , Criança , Reparo do DNA , DNA Mitocondrial/genética , Feminino , Fertilização in vitro , Conversão Gênica , Terapia Genética/ética , Terapia Genética/legislação & jurisprudência , Humanos , Masculino , Terapia de Substituição Mitocondrial , Gravidez , Diagnóstico Pré-Implantação , Segurança
19.
FEBS Lett ; 582(2): 197-202, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18078818

RESUMO

During embryo implantation, trophinin mediates cell adhesion by homophilic binding at the apical surfaces of trophectoderm and endometrium. Trophinin is expressed on the human endometrial epithelia in rare occasions. We developed hCG-coated agarose beads that mimic the physical and physiological features of an implantation-stage human blastocyst. When hCG-coated beads were applied to human endometrial epithelial cells in the presence of IL-1beta, endometrial cells acquired strong trophinin expression and the ability for apical cell adhesion with trophinin-expressing human trophoblastic cells. These results provide a mechanism for trophinin-mediated adhesion of human blastocyst to endometrium by a spatially and temporally restricted paracrine effect of hCG derived from the blastocyst.


Assuntos
Moléculas de Adesão Celular/biossíntese , Gonadotropina Coriônica/fisiologia , Endométrio/metabolismo , Interleucina-1beta/fisiologia , Adesão Celular , Endométrio/citologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Feminino , Humanos , Microscopia Imunoeletrônica
20.
BMC Biotechnol ; 8: 31, 2008 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-18366794

RESUMO

BACKGROUND: Gene targeting in nonhuman primates has the potential to produce critical animal models for translational studies related to human diseases. Successful gene targeting in fibroblasts followed by somatic cell nuclear transfer (SCNT) has been achieved in several species of large mammals but not yet in primates. Our goal was to establish the protocols necessary to achieve gene targeting in primary culture of adult rhesus macaque fibroblasts as a first step in creating nonhuman primate models of genetic disease using nuclear transfer technology. RESULTS: A primary culture of adult male fibroblasts was transfected with hTERT to overcome senescence and allow long term in vitro manipulations. Successful gene targeting of the HPRT locus in rhesus macaques was achieved by electroporating S-phase synchronized cells with a construct containing a SV40 enhancer. CONCLUSION: The cell lines reported here could be used for the production of null mutant rhesus macaque models of human genetic disease using SCNT technology. In addition, given the close evolutionary relationship and biological similarity between rhesus macaques and humans, the protocols described here may prove useful in the genetic engineering of human somatic cells.


Assuntos
Eletroporação/métodos , Fibroblastos/citologia , Fibroblastos/fisiologia , Marcação de Genes/métodos , Macaca mulatta/genética , Transfecção/métodos , Animais , Células Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA