Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 77(3): 395-406, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17522076

ABSTRACT

It has been postulated that mammalian nuclear transfer (NT) cloning efficiency is inversely correlated with donor cell differentiation status. To test this hypothesis, we compared genetically identical and increasingly differentiated donors within the myogenic lineage. Bovine male fetal muscle cells were cultured for 1-6 days in vitro. The proportion of cells displaying the following antigens was quantified by immunofluorescence microscopy: MYOD1, MYF5, PAX7, MYOG, DES, MYH, and 5-Bromo-2-deoxyuridine. Based on the antigen profile of both bulk populations and individually size-selected cells prepared for NT, donors serum-starved for 1, 4, and 5 days were classified as myogenic precursors (MPCs), myotubes (MTs), and muscle-derived fibroblasts (MFs) with purities of 92%, 85%, and 99%, respectively. Expression of the following transcripts was measured by RT-PCR in 1) cells selected for NT, 2) metaphase II oocytes, 3) NT couplets, 4) NT reconstructs, 5) NT two-cell embryos, and 6) NT blastocysts: MYOD1, MYF5, PAX7, MYOG, MYF6, ACTB, and 18S rRNA. Muscle-specific genes were silenced and remained undetectable up to the blastocyst stage, whereas housekeeping genes 18S and ACTB continued to be expressed. Differentiation status affected development to transferable embryos (118 [23%] of 520 vs. 93 [11%] of 873 vs. 66 [38%] of 174 for MPC vs. MT vs. MF, respectively, P < 0.001). However, there were no significant differences in pregnancy rate and development to weaning between the cell types (pregnancy rate: 14 [64%] of 22 vs. 8 [35%] of 23 vs. 10 [45%] of 22, and development: 4 [18%] of 22 vs. 2 [9%] of 23 vs. 3 [14%] of 22 for MPC vs. MT vs. MF, respectively).


Subject(s)
Cattle/physiology , Cloning, Organism/veterinary , Fetal Stem Cells/physiology , Muscle, Skeletal/physiology , Nuclear Transfer Techniques/veterinary , Animals , Cattle/genetics , Cell Differentiation/physiology , Cloning, Organism/methods , Desmin/chemistry , Desmin/genetics , Embryonic Development/physiology , Female , Fetal Stem Cells/cytology , Globins/chemistry , Globins/genetics , Male , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/cytology , MyoD Protein/chemistry , MyoD Protein/genetics , Myoblasts/cytology , Myoblasts/physiology , Myogenic Regulatory Factor 5/chemistry , Myogenic Regulatory Factor 5/genetics , Myogenin/chemistry , Myogenin/genetics , PAX7 Transcription Factor/chemistry , PAX7 Transcription Factor/genetics , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction/veterinary
2.
Mol Reprod Dev ; 74(5): 646-54, 2007 May.
Article in English | MEDLINE | ID: mdl-17039535

ABSTRACT

Compared to other assisted reproductive technologies, mammalian nuclear transfer (NT) cloning is inefficient in generating viable offspring. It has been postulated that nuclear reprogramming and cloning efficiency can be increased by choosing less differentiated cell types as nuclear donors. This hypothesis is mainly supported by comparative mouse cloning experiments using early blastomeres, embryonic stem (ES) cells, and terminally differentiated somatic donor cells. We have re-evaluated these comparisons, taking into account different NT procedures, the use of donor cells from different genetic backgrounds, sex, cell cycle stages, and the lack of robust statistical significance when post-blastocyst development is compared. We argue that while the reprogrammability of early blastomeres appears to be much higher than that of somatic cells, it has so far not been conclusively determined whether differentiation status affects cloning efficiency within somatic donor cell lineages.


Subject(s)
Cell Differentiation , Cloning, Organism , Nuclear Transfer Techniques , Animals , Blastomeres/cytology , Embryonic Stem Cells/cytology , Humans
3.
Cloning Stem Cells ; 8(1): 10-5, 2006.
Article in English | MEDLINE | ID: mdl-16571073

ABSTRACT

In the present study, some modifications were made to the zona-free nuclear transfer technique in the mouse in order to achieve greater efficiency. Firstly, a 1-h interval was allowed between cumulus removal and zona pellucida digestion. Secondly, acid Tyrode's was selected for zona pellucida removal, because contrary to pronase, it allows embryo survival during parthenogenic activation in the absence of calcium. Even when the exposure time to pronase was reduced to as little as 1 min or washed with fetal calf serum to inhibit the enzyme, the percentage of lysis during activation in the absence of calcium was still very high. Thirdly, electrofusion was performed at room temperature (21 degrees C), instead of 30 degrees C as in our previous experiments. Finally, embryos were cultured in groups of 12-15, instead of individually, using a "well of the wells" system during activation and culture. When compared, parthenogenic activated control embryos showed an increase in the development to blastocyst when cultured in pairs instead of individually. By the end of the experiments and using embryonic stem (ES) cells, there was a significant increase in fusion rate (1.5-fold increase) and in development to morula/blastocyst from cleaved reconstructed embryos (1.5-fold increase) when compared with the results before the modifications. A 2.4-fold increase in overall efficiency was achieved from the oocyte to morula/blastocyst stages.


Subject(s)
Cloning, Organism/methods , Nuclear Transfer Techniques , Animals , Blastomeres/cytology , Blastomeres/physiology , Calcium/pharmacology , Cell Culture Techniques , Cells, Cultured , Embryonic Development , Female , Isotonic Solutions , Mice , Morula/cytology , Morula/physiology , Ovarian Follicle/cytology , Parthenogenesis , Pronase/pharmacology , Temperature , Time Factors , Zona Pellucida
4.
Cloning Stem Cells ; 7(2): 126-38, 2005.
Article in English | MEDLINE | ID: mdl-15971986

ABSTRACT

In the present study, a zona-free nuclear transfer (NT) technique, which had been originally developed in cattle, was modified for the mouse. Steps involved in this approach include removing the zona pellucida and enucleating without a holding pipette; sticking donor cells to the cytoplast before electric pulses are applied to fuse them and culturing reconstructed embryos individually in single droplets, to prevent aggregation. Control zona-free and zona-intact embryos from mated donors showed no significant difference in development to blastocyst, but did show reduced development to term. Removal of the zona pellucida affected the response to activation by strontium in the absence of calcium as a significant proportion of zona-free control oocytes and embryos reconstructed by NT lysed during this treatment. A comparison between cumulus and ES cells as donor cells revealed significant differences in fusion efficiency (58.1 +/- 4.0%, n = 573 vs. 42.9 +/- 2.2%, n = 2064, respectively, p < 0.001), cleavage (77.2 +/- 3.4%, n = 334 vs. 40.8 +/- 2.7%, n = 903, respectively, p < 0.001) but not for development to morula/blastocyst (8.7 +/- 2.1%, n = 334 vs. 13.9 +/- 1.8%, n = 903, respectively, p < 0.1). The stage at which embryo development arrested was also affected by donor cell type. A majority of embryos reconstructed from cumulus cells arrested at two-cell stage, usually with two nuclei, whereas those reconstructed from ES cells arrested at one-cell stage, usually with two pseudo-pronuclei. After transfer of ES cell-derived NT embryos, a viable cloned mouse was produced (3.0% of transferred embryos developed to term). These observations establish that a zona-free cloning approach is possible in the mouse, although further research is required to increase the efficiency.


Subject(s)
Cloning, Organism , Nuclear Transfer Techniques , Zona Pellucida , Animals , Calcium/metabolism , Cell Fusion , Female , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Parthenogenesis
5.
Med Biol Eng Comput ; 43(1): 150-4, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15742734

ABSTRACT

Mammalian cloning by somatic nuclear transfer has great potential for developing medical applications such as biopharmaceuticals and generation of tissues for transplantation. For agricultural applications, it allows the rapid dissemination of genetic gain in livestock breeding. The maximisation of that potential requires improvements to overall cloning technology, especially with respect to increasing cloning efficiency and throughput rates in cloned embryo production. A zona-free embryo reconstruction system was developed to increase cloning throughput and ease of operation. Central to this system is a modified electrofusion procedure for nuclear transfer. Cytoplast-donor cell couplets were placed in a custom-designed 'parallel plate' electrode chamber. A 1 MHz sinusoidal AC dielectrophoresis alignment electric field of 6-10 kV m(-1) was applied for 5-10s. The couplets were then fused using 2 x 10 micros rectangular DC-field pulses (150-200 kV m(-1)), followed by application of the AC field (6-10 kV m(-1)) for another 5-10 s. Fusion was performed in hypoosmolar buffer (210 mOsm). Automated alignment of up to 20 couplets at a time has been achieved, resulting in greatly improved fusion throughput rates (2.5-fold increase) and improved fusion yields (1.3-fold increase), compared with commonly followed zona-intact protocols.


Subject(s)
Cloning, Organism/methods , Animals , Cattle , Electric Stimulation/methods , Electrophoresis/methods , Female , Male , Nuclear Transfer Techniques , Zona Pellucida
6.
Cloning Stem Cells ; 6(2): 101-10, 2004.
Article in English | MEDLINE | ID: mdl-15268783

ABSTRACT

The cloning syndrome is a continuum with the consequences of abnormal reprogramming manifest throughout gestation, the neo-natal period, and into adulthood in the cloned generation, but it does not appear to be transmitted to subsequent offspring following sexual reproduction. Most in vivo studies on bovine somatic cell cloning have focused on development during pregnancy and the neo-natal period. In this paper, we report on the viability and health of cloned cattle in adulthood. From our studies at AgResearch, we find that between weaning and 4 years of age, the annual mortality rate in cattle cloned from somatic cells is at least 8%. Although the reasons for death are variable and some potentially preventable, the main mortality factor in this period is euthanasia due to musculoskeletal abnormalities. This includes animals with severely contracted flexor tendons and those displaying chronic lameness, particularly in milking cows. In contrast, no deaths beyond weaning have so far been encountered with the offspring of clones where the oldest animals are 3 years of age. In surviving cloned cattle, blood profiles and other indicators of general physiological function such as growth rate, reproduction, rearing of offspring, and milk production are all within the normal phenotypic ranges.


Subject(s)
Cattle/genetics , Cloning, Organism/veterinary , Reproduction , Animals , Animals, Genetically Modified , Cattle/immunology , Cattle/psychology , Female , Lactation , Mortality , Obesity/pathology , Obesity/veterinary , Pregnancy , Weaning
7.
Cloning Stem Cells ; 5(1): 3-12, 2003.
Article in English | MEDLINE | ID: mdl-12713696

ABSTRACT

As the demand for cloned embryos and offspring increases, the need arises for the development of nuclear transfer procedures that are improved in both efficiency and ease of operation. Here, we describe a novel zona-free cloning method that doubles the throughput in cloned bovine embryo production over current procedures and generates viable offspring with the same efficiency. Elements of the procedure include zona-free enucleation without a holding pipette, automated fusion of 5-10 oocyte-donor cell pairs and microdrop in vitro culture. Using this system, zona-free embryos were reconstructed from five independent primary cell lines and cultured either singularly (single-IVC) or as aggregates of three (triple-IVC). Blastocysts of transferable quality were obtained at similar rates from zona-free single-IVC, triple-IVC, and control zona-intact embryos (33%, 25%, and 29%, respectively). In a direct comparison, there was no significant difference in development to live calves at term between single-IVC, triple-IVC, and zona-intact embryos derived from the same adult fibroblast line (10%, 13%, and 15%, respectively). This zona-free cloning method could be straightforward for users of conventional cloning procedures to adopt and may prove a simple, fast, and efficient alternative for nuclear cloning of other species as well.


Subject(s)
Cloning, Organism/methods , Embryo Transfer , Zona Pellucida/metabolism , Animals , Blastocyst/metabolism , Blastocyst/physiology , Cattle , Cell Line , Cell Nucleus/metabolism , Embryo, Mammalian/physiology , Female , Fertilization in Vitro , Fibroblasts/metabolism , Oocytes/metabolism
8.
Theriogenology ; 59(1): 45-59, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12499017

ABSTRACT

Several studies have shown that both quiescent and proliferating somatic donor cells can be fully reprogrammed after nuclear transfer (NT) and result in viable offspring. So far, however, no comparative study has conclusively demonstrated the relative importance of donor cell cycle stage on nuclear cloning efficiency. Here, we compare two different types of bovine fetal fibroblasts (BFFs) that were synchronized in G(0), G(1), and different phases within G(1). We show that for non-transgenic (non-TG) fibroblasts, serum starvation into G(0) results in a significantly higher percentage of viable calves at term than synchronization in early G(1) or late G(1). For transgenic fibroblasts, however, cells selected in G(1) show significantly higher development to calves at term and higher post-natal survival to weaning than cells in G(0). This suggests that it may be necessary to coordinate donor cell type and cell cycle stage to maximize overall cloning efficiency.


Subject(s)
Cattle/genetics , Cell Cycle , Cloning, Organism , Nuclear Transfer Techniques , Animals , Cattle/embryology , Embryo Transfer/veterinary , Embryonic and Fetal Development , Female , Fibroblasts/ultrastructure , G1 Phase , G2 Phase , Mitosis , Pregnancy , Resting Phase, Cell Cycle
9.
Cloning Stem Cells ; 5(4): 243-56, 2003.
Article in English | MEDLINE | ID: mdl-14733744

ABSTRACT

Over the past six years, hundreds of apparently normal calves have been cloned worldwide from bovine somatic donor cells. However, these surviving animals represent less than 5% of all cloned embryos transferred into recipient cows. Most of the remaining 95% die at various stages of development from a predictable pattern of placental and fetal abnormalities, collectively referred to as the "cloning-syndrome." The low efficiency seriously limits commercial applicability and ethical acceptance of somatic cloning and enforces the development of improved cloning methods. In this paper, we describe our current standard operating procedure (SOP) for cattle cloning using zona-free nuclear transfer. Following this SOP, the output of viable and healthy calves at weaning is about 9% of embryos transferred. Better standardization of cloning protocols across and within research groups is needed to separate technical from biological factors underlying low cloning efficiency.


Subject(s)
Cattle , Cloning, Organism/methods , Nuclear Transfer Techniques , Animals , Blastocyst/metabolism , Blastocyst/physiology , Cell Cycle , Cell Fusion/methods , Cell Nucleus/metabolism , Cells, Cultured , Embryo Transfer , Embryo, Mammalian/physiology , Female , Fertilization in Vitro , Fibroblasts/metabolism , Male , Oocytes/metabolism , Pregnancy , Zona Pellucida/physiology
10.
Proc Natl Acad Sci U S A ; 96(8): 4639-44, 1999 Apr 13.
Article in English | MEDLINE | ID: mdl-10200315

ABSTRACT

At the onset of mammalian neurogenesis, neuroepithelial (NE) cells switch from proliferative to neuron-generating divisions. Understanding the molecular basis of this switch requires the ability to distinguish between these two types of division. Here we show that in the mouse ventricular zone, expression of the mRNA of the antiproliferative gene TIS21 (PC3, BTG2) (i) starts at the onset of neurogenesis, (ii) is confined to a subpopulation of NE cells that increases in correlation with the progression of neurogenesis, and (iii) is not detected in newborn neurons. Expression of the TIS21 mRNA in the NE cells occurs transiently during the cell cycle, i.e., in the G1 phase. In contrast to the TIS21 mRNA, the TIS21 protein persists through the division of NE cells and is inherited by the neurons, where it remains detectable during neuronal migration and the initial phase of differentiation. Our observations indicate that the TIS21 gene is specifically expressed in those NE cells that, at their next division, will generate postmitotic neurons, but not in proliferating NE cells. Using TIS21 as a marker, we find that the switch from proliferative to neuron-generating divisions is initiated in single NE cells rather than in synchronized neighboring cells.


Subject(s)
Central Nervous System/embryology , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Immediate-Early Proteins/genetics , Neurons/physiology , Transcription, Genetic , Animals , Animals, Newborn , COS Cells , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division , Central Nervous System/cytology , Central Nervous System/metabolism , Epithelial Cells/cytology , Epithelial Cells/physiology , Epitopes/analysis , Immediate-Early Proteins/biosynthesis , Mice , Neurons/cytology , RNA, Messenger/analysis , RNA, Messenger/genetics , Recombinant Proteins/biosynthesis , Transfection , Tumor Suppressor Proteins
11.
EMBO J ; 18(3): 565-77, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9927416

ABSTRACT

In eukaryotic cells, tubulin polymerization must be regulated precisely during cell division and differentiation. To identify new mechanisms involved in cellular microtubule formation, we isolated an activity that suppresses microtubule nucleation in vitro. The activity was due to a small acidic polypeptide of 4.7 kDa which we named MINUS (microtubule nucleation suppressor). MINUS inhibited tau- and taxol-mediated microtubule assembly in vitro and was inactivated by dephosphorylation. The protein was purified to homogeneity from cultured neural (PC12) cells and bovine brain. Microinjection of MINUS caused a transient loss of dynamic microtubules in Vero cells. The results suggest that MINUS acts with a novel mechanism on tubulin polymerization, thus regulating microtubule formation in living cells.


Subject(s)
Microtubules/metabolism , Peptides/isolation & purification , Peptides/metabolism , Animals , Biopolymers/metabolism , Brain Chemistry , Cattle , Chlorocebus aethiops , Cytosol/chemistry , In Vitro Techniques , Microinjections , Microtubules/drug effects , Molecular Weight , PC12 Cells , Peptides/pharmacology , Rats , Tubulin/metabolism , Vero Cells
12.
Mech Dev ; 69(1-2): 71-81, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9486532

ABSTRACT

Cell differentiation often involves changes in cell polarity. In this study we show that neuroepithelial cells, the progenitors of all neurons and macroglial cells of the vertebrate central nervous system, downregulate the polarized delivery to the apical and basolateral plasma membrane domains during development. Upon infection of the neuroepithelium of mouse embryos with fowl plague virus (FPV), polarized delivery of the viral envelope hemagglutinin, an apical marker, occurred at the neural plate stage (E8), but was downregulated at the open neural tube stage (E9). Upon infection with vesicular stomatitis virus, the viral envelope G protein, a basolateral marker, showed an unpolarized delivery not only at the open neural tube stage, but already at the neural plate stage. These results show that a progressive downregulation of plasma membrane polarity of neuroepithelial cells precedes neural tube closure and the onset of neurogenesis.


Subject(s)
Cell Polarity/physiology , Membrane Glycoproteins , Nervous System/embryology , Neural Tube Defects , Neurons/cytology , Animals , Cell Membrane/virology , Down-Regulation , Embryo, Mammalian/physiology , Embryo, Mammalian/virology , Embryonic Development , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Epithelial Cells/virology , Female , Hemagglutinins, Viral/metabolism , Influenza A virus , Mice , Neurons/ultrastructure , Pregnancy , Vesicular stomatitis Indiana virus , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...