Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 29(19): 1294-1307, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32715987

ABSTRACT

Patient-specific stem cells derived from somatic cell nuclear transfer (SCNT) embryos or from induced pluripotent stem cells (iPSCs) could be used to treat various diseases with minimal immune rejection. Many studies using these cells have been conducted in rats and mice; however, there exist numerous dissimilarities between the rodents and humans limiting the clinical predictive power and experimental utility of rodent experiments alone. Nonhuman primates (NHPs) share greater homology to human than rodents in all respects, including genomics, physiology, biochemistry, and the immune system. Thus, experimental data obtained from monkey studies would be more predictive for designing an effective cell replacement therapy in humans. Unfortunately, there are few iPSC lines and even fewer SCNT lines that have been derived in NHPs, hampering broader studies in regenerative medicine. One promising potential therapy would be the replacement of dopamine neurons that are lost in Parkinson's disease. After dopamine depletion by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), the African green monkey (Chlorocebus sabaeus) shows the most complete model of Parkinsonism compared with other species and brain pathology and behavioral changes are almost identical to those in humans after accidental exposure to MPTP. Therefore, we have developed a SCNT procedure to generate multiple pluripotent stem cell lines in this species for studies of possible treatment of Parkinsonism and for comparing with cells derived from iPSCs. Using 24 female monkeys as egg donors and 7 somatic cell donor monkeys, we have derived 11 SCNT embryonic stem cell lines that expressed typical stemness genes and formed all three germ layer derivatives. We also derived two iPSC lines using an episome-mediated reprogramming factor delivery system. This report describes the process for deriving these cell lines and proving their pluripotency for differentiation into various potentially therapeutic cells.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Nuclear Transfer Techniques , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Chromosome Banding , Cloning, Organism , Culture Media , Cytogenetic Analysis , DNA/genetics , Dopaminergic Neurons/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Genotype , Humans , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Ovary/physiology , Tyrosine 3-Monooxygenase/metabolism
2.
Cell Death Dis ; 9(11): 1090, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356076

ABSTRACT

Somatic cell nuclear transfer (SCNT) or the forced expression of transcription factors can be used to generate autologous pluripotent stem cells (PSCs). Although transcriptomic and epigenomic comparisons of isogenic human NT-embryonic stem cells (NT-ESCs) and induced PSCs (iPSCs) in the undifferentiated state have been reported, their functional similarities and differentiation potentials have not been fully elucidated. Our study showed that NT-ESCs and iPSCs derived from the same donors generally displayed similar in vitro commitment capacity toward three germ layer lineages as well as proliferative activity and clonogenic capacity. However, the maturation capacity of NT-ESC-derived hematopoietic progenitors was significantly greater than the corresponding capacity of isogenic iPSC-derived progenitors. Additionally, donor-dependent variations in hematopoietic specification and commitment capacity were observed. Transcriptome and methylome analyses in undifferentiated NT-ESCs and iPSCs revealed a set of genes that may influence variations in hematopoietic commitment and maturation between PSC lines derived using different reprogramming methods. Here, we suggest that genetically identical iPSCs and NT-ESCs could be functionally unequal due to differential transcription and methylation levels acquired during reprogramming. Our proof-of-concept study indicates that reprogramming mechanisms and genetic background could contribute to diverse functionalities between PSCs.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Hematopoietic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Alveolar Epithelial Cells/physiology , Cell Line , Cell Proliferation , Colony-Forming Units Assay , DNA Methylation/physiology , Germ Layers/physiology , Human Embryonic Stem Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Nuclear Transfer Techniques , Transcription, Genetic/physiology , Transcriptome
3.
Dev Reprod ; 21(4): 425-434, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29359202

ABSTRACT

Polyploidy is occurred by the process of endomitosis or cell fusion and usually represent terminally differentiated stage. Their effects on the developmental process were mainly investigated in the amphibian and fishes, and only observed in some rodents as mammalian model. Recently, we have established tetraploidy somatic cell nuclear transfer-derived human embryonic stem cells (SCNT-hESCs) and examined whether it could be available as a research model for the polyploidy cells existed in the human tissues. Two tetraploid hESC lines were artificially acquired by reintroduction of remained 1st polar body during the establishment of SCNT-hESC using MII oocytes obtained from female donors and dermal fibroblasts (DFB) from a 35-year-old adult male. These tetraploid SCNT-hESC lines (CHA-NT1 and CHA-NT3) were identified by the cytogenetic genotyping (91, XXXY,-6, t[2:6] / 92,XXXY,-12,+20) and have shown of indefinite proliferation, but slow speed when compared to euploid SCNT-hESCs. Using the eight Short Tendem Repeat (STR) markers, it was confirmed that both CHA-NT1 and CHA-NT3 lines contain both nuclear and oocyte donor genotypes. These hESCs expressed pluripotency markers and their embryoid bodies (EB) also expressed markers of the three embryonic germ layers and formed teratoma after transplantation into immune deficient mice. This study showed that tetraploidy does not affect the activities of proliferation and differentiation in SCNT-hESC. Therefore, tetraploid hESC lines established after SCNT procedure could be differentiated into various types of cells and could be an useful model for the study of the polyploidy cells in the tissues.

4.
BMB Rep ; 49(4): 197-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26996342

ABSTRACT

Although three different research groups have reported successful derivations of human somatic cell nuclear transfer-derived embryonic stem cell (SCNT-ESC) lines using fetal, neonatal and adult fibroblasts, the extremely poor development of cloned embryos has hindered its potential applications in regenerative medicine. Recently, however, our group discovered that the severe methylation of lysine 9 in Histone H3 in a human somatic cell genome was a major SCNT reprogramming barrier, and the overexpression of KDM4A, a H3K9me3 demethylase, significantly improved the blastocyst formation of SCNT embryos. In particular, by applying this new approach, we were able to produce multiple SCNT-ES cell lines using oocytes obtained from donors whose eggs previously failed to develop to the blastocyst stage. Moreover, the success rate was closer to 25%, which is comparable to that of IVF embryos, so that our new human SCNT method seems to be a practical approach to establishing a pluripotent stem cell bank for the general public as well as for individual patients. [BMB Reports 2016; 49(4): 197-198].


Subject(s)
Nuclear Transfer Techniques , Pluripotent Stem Cells/cytology , Tissue Banks , Fertilization in Vitro , Genome, Human , Human Embryonic Stem Cells/cytology , Humans , Stem Cell Transplantation
5.
Cell Stem Cell ; 17(6): 758-766, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26526725

ABSTRACT

The extremely low efficiency of human embryonic stem cell (hESC) derivation using somatic cell nuclear transfer (SCNT) limits its potential application. Blastocyst formation from human SCNT embryos occurs at a low rate and with only some oocyte donors. We previously showed in mice that reduction of histone H3 lysine 9 trimethylation (H3K9me3) through ectopic expression of the H3K9me3 demethylase Kdm4d greatly improves SCNT embryo development. Here we show that overexpression of a related H3K9me3 demethylase KDM4A improves human SCNT, and that, as in mice, H3K9me3 in the human somatic cell genome is an SCNT reprogramming barrier. Overexpression of KDM4A significantly improves the blastocyst formation rate in human SCNT embryos by facilitating transcriptional reprogramming, allowing efficient derivation of SCNT-derived ESCs using adult Age-related Macular Degeneration (AMD) patient somatic nuclei donors. This conserved mechanistic insight has potential applications for improving SCNT in a variety of contexts, including regenerative medicine.


Subject(s)
Cellular Reprogramming , Gene Expression Regulation, Enzymologic , Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Oocytes/cytology , Pluripotent Stem Cells/cytology , Animals , Blastocyst/cytology , Cell Nucleus/metabolism , Cumulus Cells/cytology , Female , Heterochromatin/metabolism , Histones/chemistry , Humans , Lysine/chemistry , Macular Degeneration/metabolism , Mice , Nuclear Transfer Techniques , RNA, Messenger/metabolism , Transcription, Genetic
6.
Cell Stem Cell ; 14(6): 777-80, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24746675

ABSTRACT

Derivation of patient-specific human pluripotent stem cells via somatic cell nuclear transfer (SCNT) has the potential for applications in a range of therapeutic contexts. However, successful SCNT with human cells has proved challenging to achieve, and thus far has only been reported with fetal or infant somatic cells. In this study, we describe the application of a recently developed methodology for the generation of human ESCs via SCNT using dermal fibroblasts from 35- and 75-year-old males. Our study therefore demonstrates the applicability of SCNT for adult human cells and supports further investigation of SCNT as a strategy for regenerative medicine.


Subject(s)
Adult Stem Cells/cytology , Nuclear Transfer Techniques , Pluripotent Stem Cells/cytology , Adult , Aged , Dermis/cytology , Fibroblasts/cytology , Humans , Male
8.
Methods Mol Biol ; 348: 111-24, 2006.
Article in English | MEDLINE | ID: mdl-16988375

ABSTRACT

Cloning by somatic cell nuclear transfer is a complex procedure that is dependent on correct interactions between oocyte and donor cell genome. These interactions require minimal insult to either the oocyte or the transplanted nucleus. Available data also indicate that reprogramming the donor cell genome may be slow, so that the cloned embryo expresses genes typical of the donor cell, and thus has different characteristics from normal embryos. Procedures that minimize damage to the donor genome and that address the unique characteristics of the cloned construct should enhance the efficacy of the method.


Subject(s)
Cloning, Organism/standards , Embryo Culture Techniques/methods , Epigenesis, Genetic , Fertilization in Vitro , Mice , Nuclear Transfer Techniques , Animals , Cells, Cultured , Cloning, Organism/methods , Genomic Imprinting
9.
Biol Reprod ; 72(3): 612-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15537860

ABSTRACT

Interactions between sperm and egg are required to maintain embryo viability and cellular integrity. Differential transcriptional activities and epigenetic differences that include genomic imprinting provide mechanisms by which complementary parental genome functions support early embryogenesis. We previously showed that cytofragmentation can be influenced by the specific combination of maternal and paternal genotypes. Using maternal pronuclear transfer in mouse embryos, we examined the cellular basis for the maternal genotype effect. We found that the maternal genotype effect is predominantly controlled by the maternal pronucleus, with a lesser role played by the ooplasm. This effect of the maternal pronucleus is sensitive to alpha-amanitin treatment. The effect of the maternal component of the embryonic genome on cytofragmentation constitutes the earliest known effect of the embryonic genome on mammalian embryo phenotype. The results also indicate that clinical procedures seeking to define or manipulate oocyte quality in humans should take into account early effects of the embryonic genome, particularly the maternal genome.


Subject(s)
Apoptosis/physiology , Blastomeres/physiology , Cell Nucleus/physiology , Embryonic Development/physiology , Ovum/physiology , Animals , Apoptosis/genetics , Blastomeres/cytology , Cell Nucleus/genetics , Cytoplasm/physiology , Embryonic Development/genetics , Epigenesis, Genetic/physiology , Female , Genotype , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Ovum/cytology , Pregnancy , Spermatozoa/physiology
10.
Dev Biol ; 266(1): 62-75, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14729478

ABSTRACT

H1 linker histones (H1s) are key regulators of chromatin structure and function. The functions of different H1s during early embryogenesis, and mechanisms regulating their associations with chromatin are largely unknown. The developmental transitions of H1s during oocyte growth and maturation, fertilization and early embryogenesis, and in cloned embryos were examined. Oocyte-specific H1FOO, but not somatic H1s, associated with chromatin in oocytes (growing, GV-stage, and MII-arrested), pronuclei, and polar bodies. H1FOO associated with sperm or somatic cell chromatin within 5 min of intracytoplasmic sperm injection (ICSI) or somatic cell nuclear transfer (SCNT), and completely replaced somatic H1s by 60 min. The switching from somatic H1s to H1FOO following SCNT was developmentally regulated. H1FOO was replaced by somatic H1s during the late two- and four-cell stages. H1FOO association with chromatin can occur in the presence of a nuclear envelope and independently of pronucleus formation, is regulated by factors associated with the spindle, and is likely an active process. All SCNT constructs recapitulated the normal sequence of H1 transitions, indicating that this alone does not signify a high developmental potential. A paucity of all known H1s in two-cell embryos may contribute to precocious gene transcription in fertilized embryos, and the elaboration of somatic cell characteristics in cloned embryos.


Subject(s)
Cell Nucleus , Fertilization , Histones/physiology , Sperm Injections, Intracytoplasmic , Animals , Embryonic and Fetal Development , Female , Fluorescent Antibody Technique , Mice , Oocytes/physiology
11.
Biol Reprod ; 70(4): 1162-70, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14681201

ABSTRACT

Previous studies revealed that oocytes of different genetic strains (e.g, C57BL/6 and DBA/2) modify maternal and paternal pronuclei differently, affecting early preimplantation development. To determine whether these strain-dependent effects would also apply to oocyte modifications of somatic cell nuclei introduced during cloning procedures, we compared the efficiency of development of parthenogenetic and cloned embryos made with DBA/2, C57BL/6, and (B6D2)F1 oocytes. Our results reveal significant differences in the ability of oocytes of different genetic backgrounds to support parthenogenetic development in different culture media. Additionally, our results reveal oocyte strain-dependent differences in the ability to support cloned embryo development beyond what can be accounted for on the basis of differences in parthenogenesis. Thus, the previously documented differences in oocyte-directed parental genome modification are accompanied in the same strains by differences in the ability of oocytes to modify somatic cell nuclei and support clonal development, raising the possibility that these oocyte functions may be mediated by related mechanisms. These results provide a genetic basis for further studies seeking to identify specific genes that determine oocyte phenotype, as well as genes that determine the success of nuclear reprogramming and clonal development.


Subject(s)
Cloning, Organism , Genetic Variation , Oocytes/physiology , Parthenogenesis , Animals , Carrier Proteins/metabolism , Embryonic and Fetal Development , Female , Genotype , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Phenotype , Tissue Donors
12.
Biol Reprod ; 69(3): 902-14, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12748125

ABSTRACT

Cloning by somatic cell nuclear transfer requires that epigenetic information possessed by the donor nucleus be reprogrammed to an embryonic state. Little is known, however, about this remodeling process, including when it occurs, its efficiency, and how well epigenetic markings characteristic of normal development are maintained. Examining the fate of epigenetic information associated with imprinted genes during clonal development offers one means of addressing these questions. We examined transcript abundance, allele specificity of imprinted gene expression, and parental allele-specific DNA methylation in cloned mouse blastocysts. Striking disruptions were seen in total transcript abundance and allele specificity of expression for five imprinted genes. Only 4% of clones recapitulated a blastocyst mode of expression for all five genes. Cloned embryos also exhibited extensive loss of allele-specific DNA methylation at the imprinting control regions of the H19 and Snprn genes. Thus, epigenetic errors arise very early in clonal development in the majority of embryos, indicating that reprogramming is inefficient and that some epigenetic information may be lost.


Subject(s)
Blastocyst/metabolism , Cloning, Organism , Epigenesis, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genomic Imprinting , Allelic Imbalance/genetics , Animals , DNA Methylation , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Nuclear Transfer Techniques
13.
Biol Reprod ; 69(1): 146-53, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12606374

ABSTRACT

Cloning by somatic cell nuclear transfer is inefficient. This is evident in the significant attrition in the number of surviving cloned offspring at virtually all stages of embryonic and fetal development. We find that cloned preimplantation mouse embryos aberrantly express the somatic form of the Dnmt1 DNA (cytosine-5) methyltransferase, the expression of which is normally prevented by a posttranscriptional mechanism. Additionally, the maternal oocyte-derived Dnmt1o isoform undergoes little or none of its expected translocation to embryonic nuclei at the eight-cell stage. Such defects in the regulation of Dnmt1s and Dnmt1o expression and cytoplasmic-nuclear trafficking may prevent clones from completing essential early developmental events. Furthermore, aberrant Dnmt1 localization and expression may contribute to the defects in DNA methylation and the developmental abnormalities seen in cloned mammals.


Subject(s)
Blastocyst/enzymology , Cloning, Organism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Animals , Cell Nucleus/enzymology , Cleavage Stage, Ovum/enzymology , Cytoplasm/enzymology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Female , Immunohistochemistry , Male , Mice , Morula/enzymology , Mosaicism , Nuclear Transfer Techniques , Pregnancy
14.
Biol Reprod ; 69(1): 48-56, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12606377

ABSTRACT

Cloning by somatic cell nuclear transfer requires silencing of the donor cell gene expression program and the initiation of the embryonic gene expression program (nuclear reprogramming). Failure to silence the donor cell program could lead to altered embryonic phenotypes. Cloned mouse embryos produced using myoblast nuclei fail to thrive in standard embryo culture media but flourish in somatic cell culture media favored by the donor myoblasts themselves, forming blastocysts at a significant rate, with robust morphologies, high total cell number, and a normal allocation of cells to the inner cell mass in most embryos. Myoblast cloned embryos continue expressing the GLUT4 glucose transporter, which is typically expressed in muscle but not in preimplantation stage embryos. Myoblast clones also exhibit precocious enrichment of GLUT1 at the cell surface. Both myoblast and cumulus cell cloned embryos exhibit enhanced rates of glucose uptake. These observations indicate that silencing of the donor cell genome during cloning either is incomplete or occurs progressively over the course of preimplantation development. As a result, cloned embryos initially exhibit many somatic cell-like characteristics. Tetraploid constructs, which possess a transplanted somatic cell genome plus the oocyte-derived chromosomes, exhibit a more embryonic-like pattern of gene expression and culture preference. We conclude that preimplantation stage cloned embryos have profoundly altered characteristics that are donor cell type specific and that exposure of cloned embryos to standard embryo culture conditions may lead to disruptions in basic homeostasis and inhibition of a range of essential processes including further nuclear reprogramming, contributing to cloned embryo demise.


Subject(s)
Cloning, Organism/methods , Muscle Proteins , Myoblasts/cytology , Nuclear Transfer Techniques , Animals , Blastocyst/cytology , Blastocyst/metabolism , Culture Media , Gene Expression , Gene Silencing , Glucose/metabolism , Glucose Transporter Type 1 , Glucose Transporter Type 4 , In Vitro Techniques , Mice , Monosaccharide Transport Proteins/genetics , Myoblasts/metabolism , Phenotype
15.
Biol Reprod ; 66(4): 1178-84, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11906939

ABSTRACT

Cloning by somatic cell nuclear transfer is critically dependent upon early events that occur immediately after nuclear transfer, and possibly additional events that occur in the cleaving embryo. Embryo culture conditions have not been optimized for cloned embryos, and the effects of culture conditions on these early events and the successful initiation of clonal development have not been examined. To evaluate the possible effect of culture conditions on early cloned embryo development, we have compared a number of different culture media, either singly or in sequential combinations, for their ability to support preimplantation development of clones produced using cumulus cell nuclei. We find that glucose is beneficial during the 1-cell stage when CZB medium is employed. We also find that potassium simplex optimized medium (KSOM), which is optimized to support efficient early cleavage divisions in mouse embryos, does not support development during the 1-cell or 2-cell stages in the cloned embryos as well as other media. Glucose-supplemented CZB medium (CZB-G) supports initial development to the 2-cell stage very well, but does not support later cleavage stages as well as Whittten medium or KSOM. Culturing cloned embryos either entirely in Whitten medium or initially in Whittens medium and then changing to KSOM at the late 4-cell/early 8-cell stage produces consistent production of blastocysts at a greater frequency than using CZB-G medium alone. The combination of Whitten medium followed by KSOM resulted in an increased number of cells per blastocyst. Because normal embryos do not require glucose during the early cleavage stages and develop efficiently in all of the media employed, these results reveal unusual culture medium requirements that are indicative of altered physiology and metabolism in the cloned embryos. The relevance of this to understanding the kinetics and mechanisms of nuclear reprogramming and to the eventual improvement of the overall success in cloning is discussed.


Subject(s)
Cell Nucleus/physiology , Cloning, Organism , Culture Media , Cytoplasm/physiology , Embryonic Development , Animals , Blastocyst/physiology , Culture Techniques , Dimethyl Sulfoxide/pharmacology , Embryonic and Fetal Development/drug effects , Female , Glucose/administration & dosage , Mice , Mice, Inbred C57BL , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...