US20070033664A1 - Quiescent cell populations for nuclear transfer - Google Patents

Quiescent cell populations for nuclear transfer Download PDF

Info

Publication number
US20070033664A1
US20070033664A1 US11/543,786 US54378606A US2007033664A1 US 20070033664 A1 US20070033664 A1 US 20070033664A1 US 54378606 A US54378606 A US 54378606A US 2007033664 A1 US2007033664 A1 US 2007033664A1
Authority
US
United States
Prior art keywords
cell
embryo
cells
embryos
recipient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/543,786
Inventor
Keith Campbell
Ian Wilmut
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Roslin Institute Edinburgh
Original Assignee
Roslin Institute Edinburgh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roslin Institute Edinburgh filed Critical Roslin Institute Edinburgh
Priority to US11/543,786 priority Critical patent/US20070033664A1/en
Publication of US20070033664A1 publication Critical patent/US20070033664A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/873Techniques for producing new embryos, e.g. nuclear transfer, manipulation of totipotent cells or production of chimeric embryos
    • C12N15/877Techniques for producing new mammalian cloned embryos
    • C12N15/8771Bovine embryos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/873Techniques for producing new embryos, e.g. nuclear transfer, manipulation of totipotent cells or production of chimeric embryos
    • C12N15/877Techniques for producing new mammalian cloned embryos
    • C12N15/8773Ovine embryos

Definitions

  • This invention relates to the generation of animals including but not being limited to genetically selected and/or modified animals.
  • the reconstruction of mammalian embryos by the transfer of a nucleus from a donor embryo to an enucleated oocyte or one cell zygote allows the production of genetically identical individuals. This has clear advantages for both research (i.e. as biological controls) and also in commercial applications (i.e. multiplication of genetically valuable livestock, uniformity of meat products, animal management).
  • One problem with the use of early embryos as nuclear donors is that the number of offspring which can be produced from a single embryo is limited both by the number of cells (embryos at the 32-64 cell stage are the most widely used in farm animal species) and the efficiency of the nuclear transfer protocol.
  • ES Embryonic Stem
  • ES cell lines There are several alternatives to the use of ES cell lines; one of these is to search for other cell populations which are able to promote development when used for nuclear transfer.
  • Primordial Germ Cells offer a suitable candidate; however no development to term has yet been reported.
  • Cell lines established from early embryos have been suggested; although development has been reported from early passage cells in the sheep (Campbell et al., Therio 43 181 (1995)) on prolonged culture, no development was obtained using conventional nuclear transfer protocols (Campbell et al., J. Abstract Series (5) 31 (1995)).
  • a totipotent cell can direct the development of a whole animal (when constructing embryos by nuclear transfer from a donor cell into a recipient cell, such as an enucleated oocyte, it is the nucleus of the donor cell which is totipotent). This includes directing the development of extra-embryonic lineages, i.e. the placenta. In this definition, a fertilised zygote and in some species individual blastomeres are also totipotent. In contradistinction, a pluripotent or multipotent cell (i.e. an embryonic stem cell) type has been defined as one which can form all tissues in the conceptus/offspring after injection into the blastocoele cavity.
  • quiescent cells that is to say cells which are not actively proliferating by means of the cell cycle
  • quiescent cells can advantageously be used as nuclear donors in the reconstitution of an animal embryo. Such embryos may then be allowed to develop to term. It seems that changes in the donor nucleus which are observed after embryo reconstruction and which are required for efficient nuclear transfer can be induced in the nuclei of cells prior to their use as nuclear donors by causing them to enter the quiescent state. This fact has been exploited in the present application.
  • a method of reconstituting an animal embryo comprising transferring the nucleus of a quiescent donor cell into a suitable recipient cell.
  • the invention is applicable to all animals, including birds, such as domestic fowl, amphibian species and fish species. In practice, however, it will be to non-human animals, especially (non-human) mammals, particularly placental mammals, that the greatest commercially useful applicability is presently envisaged. It is with ungulates, particularly economically important ungulates such as cattle, sheep, goats, water buffalo, camels and pigs that the invention is likely to be most useful, both as a means for cloning animals and as a means for generating transgenic or genetically modified animals. It should also be noted that the invention is also likely to be applicable to other economically important animal species such as, for example, horses, llamas or rodents e.g. rats or mice, or rabbits.
  • Transgenic animals may be produced from genetically altered donor cells.
  • the overall procedure has a number of advantages over conventional procedures for the production of transgenic (i.e. genetically modified) animals which may be summarised as follows:
  • transgenic in relation to animals, should not be taken to be limited to referring to animals containing in their germ line one or more genes from another species, although many transgenic animals will contain such a gene or genes. Rather, the term refers more broadly to any animal whose germ line has been the subject of technical intervention by recombinant DNA technology. So, for example, an animal in whose germ line an endogenous gene has been deleted, duplicated, activated or modified is a transgenic animal for the purposes of this invention as much as an animal to whose germ line an exogenous DNA sequence has been added.
  • the donor nucleus is genetically modified.
  • the donor nucleus may contain one or more transgenes and the genetic modification may take place prior to nuclear transfer and embryo reconstitution.
  • micro-injection analogous to injection into the male or female pronucleus of a zygote, may be used as a method of genetic modification, the invention is not limited to that methodology: mass transformation or transfection techniques can also be used e.g. electroporation, viral transfection or lipofection.
  • a nucleus is transferred from a quiescent donor cell to a recipient cell.
  • the use of this method is not restricted to a particular donor cell type. All cells of normal karyotype, including embryonic, foetal and adult somatic cells, which can be induced to enter quiescence or exist in a quiescent state in vivo may prove totipotent using this technology.
  • the invention therefore contemplates the use of an at least partially differentiated cell, including a fully differentiated cell.
  • Donor cells may be, but do not have to be, in culture.
  • TNT4 embryo-derived ovine cell line
  • OME ovine mammary epithelial cell derived cell line
  • SECL epithelial-like cell line derived from a 9-day old sheep embryo
  • donor cells are quiescent, which is to say that they are not actively proliferating by means of the mitotic cell cycle.
  • the mitotic cell cycle has four distinct phases, G1, S, G2 and M.
  • the beginning event in the cell cycle, called start takes place in the G1 phase and has a unique function.
  • the decision or commitment to undergo another cell cycle is made at start.
  • Once a cell has passed through start it passes through the remainder of the G1 phase, which is the pre-DNA synthesis phase.
  • the second stage, the S phase is when DNA synthesis takes place. This is followed by the G2 phase, which is the period between DNA synthesis and mitosis. Mitosis itself occurs at the M phase.
  • Quiescent cells which include cells in which quiescence has been induced as well as those cells which are naturally quiescent, such as certain fully differentiated cells
  • the nuclei of quiescent G0 cells have a diploid DNA content.
  • Cultured cells can be induced to enter the quiescent state by various methods including chemical treatments, nutrient deprivation, growth inhibition or manipulation of gene expression. Presently the reduction of serum levels in the culture medium has been used successfully to induce quiescence in both ovine and bovine cell lines. In this situation, the cells exit the growth cycle during the G1 phase and arrest, as explained above, in the so-called G0 stage. Such cells can remain in this state for several days (possibly longer depending upon the cell) until re-stimulated when they re-enter the growth cycle.
  • Quiescent cells arrested in the G0 state are diploid. The G0 state is the point in the cell cycle from which cells are able to differentiate.
  • the recipient cell to which the nucleus from the donor cell is transferred may be an oocyte or another suitable cell.
  • Recipient cells at a variety of different stages of development may be used, from oocytes at metaphase I through metaphase II, to zygotes and two-cell embryos. Each has its advantages and disadvantages.
  • the use of fertilized eggs ensures efficient activation whereas parthenogenetic activation is required with oocytes (see below).
  • Another mechanism that may favour the use of cleavage-stage embryos in some species is the extent to which reprogramming of gene expression is required. Transcription is initiated during the second cell cycle in the mouse and no major changes in the nature of the proteins being synthesised are revealed by two-dimensional electrophoresis until the blastocyst stage (Howlett & Bolton J. Embryol. Exp. Morphol. 87 175-206 (1985)). In most cases, though, the recipient cells will be oocytes.
  • the recipient be enucleate. While it has been generally assumed that enucleation of recipient oocytes in nuclear transfer procedures is essential, there is no published experimental confirmation of this judgement.
  • the original procedure described for ungulates involved splitting the cell into two halves, one of which was likely to be enucleated (Willadsen Nature 320 (6) 63-65 (1986)). This procedure has the disadvantage that the other unknown half will still have the metaphase apparatus and that the reduction in volume of the cytoplasm is believed to accelerate the pattern of differentiation of the new embryo (Eviskov et al., Development 109 322-328 (1990)).
  • recipient host cells to which the donor cell nucleus is transferred is an enucleated metaphase II oocyte, an enucleated unactivated oocyte or an enucleated preactivated oocyte. At least where the recipient is an enucleated metaphase II oocyte, activation may take place at the time of transfer. Alternatively, at least where the recipient is an enucleated unactivated metaphase II oocyte, activation may take place subsequently. As described above enucleation may be achieved physically, by actual removal of the nucleus, pro-nuclei or metaphase plate (depending on the recipient cell), or functionally, such as by the application of ultraviolet radiation or another enucleating influence.
  • cytoplast Enucleated oocyte recipients
  • the “GOAT” G0/G1 Activation and Transfer—a MII (metaphase II) oocyte at the time of activation (Campbell et al., Biol. Reprod. 49 933-942 (1993).
  • nucleus of the former it is necessary for the nucleus of the former to be transferred to the latter. Most conveniently, nuclear transfer is effected by fusion.
  • fusion is commonly achieved by the same electrical stimulation that is used to induce parthogenetic activation (Willadsen Nature 320 (6) 63-65 (1986), Prather et al., Biol. Reprod. 37 859-866 (1987)).
  • Sendai virus induces fusion in a proportion of cases, but is not sufficiently reliable for routine application (Willadsen Nature 320 (6) 63-65 (1986)).
  • cell-cell fusion is a preferred method of effecting nuclear transfer, it is not the only method that can be used.
  • suitable techniques include microinjection (Ritchie and Campbell, J. Reproduction and Fertility Abstract Series No. 15, p60).
  • parthenogenetic activation Before or (preferably) after nuclear transfer (or, in some instances at least, concomitantly with it), it is generally necessary to stimulate the recipient cell into development by parthenogenetic activation, at least if the cell is an oocyte.
  • parthogenetic activation is more complicated than had been imagined. It had been assumed that activation is an all-or-none phenomenon and that the large number of treatments able to induce formation of a pronucleus were all causing “activation”.
  • exposure of rabbit oocytes to repeated electrical pulses revealed that only selection of an appropriate series of pulses and control of the Ca 2+ was able to promote development of diploidized oocytes to mid-gestation (Ozil Development 109 117-127 (1990)).
  • the interval between pulses in the rabbit is approximately 4 minutes (Ozil Development 109 117-127 (1990)), and in the mouse 10 to 20 minutes (Cutbertson & Cobbold Nature 316 541-542 (1985)), while there are preliminary observations in the cow that the interval is approximately 20 to 30 minutes (Robl et al., in Symposium on cloning Mammals by Nuclear Transplantation (Seidel ed.), Colorado State University, 24-27 (1992)). In most published experiments activation was induced with a single electrical pulse, but new observations suggest that the proportion of reconstituted embryos that develop is increased by exposure to several pulses (Collas & Robl Biol. Reprod. 43 877-884 (1990)). In any individual case, routine adjustments may be made to optimise the number of pulses, the field strength and duration of the pulses and the calcium concentration of the medium.
  • a reconstituted animal embryo prepared by a method as described previously.
  • a method for preparing an animal comprising:
  • Step (a) has been described in depth above.
  • step (b) in the method of this aspect of the invention is to cause an animal to develop to term from the embryo. This may be done directly or indirectly. In direct development, the reconstituted embryo from step (a) is simply allowed to develop without further intervention beyond any that may be necessary to allow the development to take place. In indirect development, however, the embryo may be further manipulated before full development takes place. For example, the embryo may be split and the cells clonally expanded, for the purpose of improving yield.
  • each embryo that does develop itself can be used as a nuclear donor, such as, for example at the morula or 32-64 cell stage; alternatively, inner cell mass cells can be used at the blastocyst stage. Embryos derived from these subsequent transfers could themselves also be used as potential nuclear donors to further increase efficiency.
  • each developing embryo may be multiplied in this way by the efficiency of the nuclear transfer process.
  • the degree of enhancement likely to be achieved depends upon the cell type. In sheep, it is readily possible to obtain 55% blastocyst stage embryos by transfer of a single blastomere from a 16 cell embryo to a preactivated “Universal Recipient” oocyte. So it is reasonable to hypothesise that each embryo developed from a single cell could give rise to eight at the 16 cell stage. Although these figures are just a rough guide, it is clear that at later developmental stages the extent of benefit would depend on the efficiency of the process at that stage.
  • a new cell line to act as a source of nuclear donor cells could be produced from embryos formed according to the preceding description or the resulting foetuses or adults.
  • a chimeric animal formed from cells derived from a naturally formed embryo and an embryo reconstructed by nuclear transfer.
  • a chimera can be formed by taking a proportion of cells of the natural embryo and a proportion of the cells of the reconstructed embryo at any stage up to the blastocyst stage and forming a new embryo by aggregation or injection.
  • the proportion of cells may be in the ratio of 50:50 or another suitable ratio to achieve the formation of an embryo which develops to term. The presence of normal cells in these circumstances is thought to assist in rescuing the reconstructed embryo and allowing successful development to term and a live birth.
  • the reconstituted embryo may be cultured, in viva or in vitro to blastocyst.
  • the embryo in order to protect the embryo it should preferably be embedded in a protective medium such as agar before transfer and then dissected from the agar after recovery from the temporary recipient.
  • a protective medium such as agar before transfer and then dissected from the agar after recovery from the temporary recipient.
  • the function of the protective agar or other medium is twofold: first, it acts as a structural aid for the embryo by holding the zona pellucida together; and secondly it acts as barrier to cells of the recipient animal's immune system.
  • the embryo may be screened for suitability for development to term. Typically, this will be done where the embryo is transgenic and screening and selection for stable integrants has been carried out. Screening for non-transgenic genetic markers may also be carried out at this stage. However, because the method of the invention allows for screening of donors at an earlier stage, that will generally be preferred.
  • blastocyst embryo After screening, if screening has taken place, the blastocyst embryo is allowed to develop to term. This will generally be in vivo. If development up to blastocyst has taken place in vitro, then transfer into the final recipient animal takes place at this stage. If blastocyst development has taken place in vivo, although in principle the blastocyst can be allowed to develop to term in the pre-blastocyst host, in practice the blastocyst will usually be removed from the (temporary). pre-blastocyst recipient and, after dissection from the protective medium, will be transferred to the (permanent) post-blastocyst recipient.
  • animals may be bred from the animal prepared by the preceding steps. In this way, an animal may be used to establish a herd or flock of animals having the desired genetic characteristic(s).
  • Oocytes can be obtained by (i) in vitro maturation of slaughterhouse material, or from transvaginal follicle puncture; (ii) in vivo maturation and surgically recovery; or (iii) any other suitable procedure. All in vivo matured oocytes should be harvested by flushing from the oviduct in calcium magnesium free phosphate buffered saline (PBS) containing 1.0% foetal calf serum (FCS). In vitro matured oocytes are harvested and transferred to calcium free M2 (Whittingham and Wales Aust. J. Biol. Sci. 22 1065-1068 (1969)) containing 1.0% FCS.
  • PBS calcium magnesium free phosphate buffered saline
  • FCS foetal calf serum
  • Oocytes are denuded of cumulus, cells and enucleated as previously described (Campbell et al., Biol. Reprod. 49 933-942 (1993) and Biol. Reprod. 50 1385-1393 (1994)) with the exception that calcium free medium is used for all procedures. Fusion procedures are modifications of those previously reported (Campbell et al., 1993, 1994 loc cit) and are as described in the relevant section below, alternatively the nucleus may be introduced by injection of the donor cell into the enucleated oocyte (Ritchie & Campbell, J. Reprod. Fertil. Abstract Series (5) 60 (1995)). The timing of these events is dependent upon the species, the following two protocols outline the use of in vivo matured ovine and in vitro matured bovine oocytes.
  • Unfertilised metaphase II oocytes were recovered by flushing from the oviduct at 24-29 hours after GnRH injection using Dulbeccols phosphate buffered saline containing 1.0% foetal calf serum (FCS) maintained at 37° C. until use.
  • FCS foetal calf serum
  • Recovered oocytes were maintained at 37° C., washed in PBS 1.0% FCS and transferred to calcium free M2 medium containing 10% Foetal Calf Serum (FCS), at 37° C.
  • FCS Foetal Calf Serum
  • (enucleation) oocytes were placed in calcium free M2 containing 10% FCS, 7.5 ⁇ g/ml cytochalasin B (Sigma) and 5.0 ⁇ g/ml Hoechst 33342 (Sigma) at 37° C. for 20 minutes.
  • a small amount of cytoplasm from directly beneath the 1st polar body was then aspirated using a 20 ⁇ M glass pipette. Enucleation was confirmed by exposing the aspirated portion of cytoplasm to UV light and checking for the presence of a metaphase plate.
  • a single cell was placed into contact with the oocyte by using a glass pipette to transfer the cell through the hole previously made in the zona pellucida.
  • the cytoplast/cell couplet was then transferred into the fusion chamber in 200 ⁇ l of 0.3M mannitol in distilled water and manually aligned between the electrodes.
  • An AC pulse of 5V was applied for 3 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 ⁇ secs.
  • the couplets were then washed in calcium free M2, 10% FCS at 37° C. and incubated in the same medium under oil at 37° C. 5% CO 2 .
  • oocytes were placed between two parallel electrodes in 200 ⁇ l of 0.3M mannitol, 0.1 mM MgSO 4 . 0.001 mM CaCl 2 in distilled water (Willadsen, Nature 320 63-65 (1986)). Activation was induced by application of 1 DC pulse of 1.25 kV/cm for 80 ⁇ s.
  • manipulated embryos were treated in a similar manner with the addition that the contact surface between the enucleated oocyte and the cell was arranged parallel to the electrodes. Fusion was induced by application of an AC current of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 ⁇ s.
  • In vitro techniques may also be suitable in place of a temporary recipient ewe to achieve development of the embryo to the blastocyst stage.
  • Ovaries were obtained from a local abattoir and maintained at 28-32° C. during transport to the laboratory.
  • Cumulus oocyte complexes COC's
  • COC's Cumulus oocyte complexes
  • the universal containers were placed into a warmed chamber (35° C.) and the follicular material allowed to settle for 10-15 minutes before pouring off three quarters of the supernatant.
  • the remaining follicular material was diluted with an equal volume of dissection medium (TCM 199 with Earles salts (Gibco), 75.0 mg/l kanamycin, 30.0 mM Hepes, pH 7.4, osmolarity 280 mOsmols/Kg H 2 O) supplemented with 10% bovine serum, transferred into an 85 mm petri dish and searched for COC's under a dissecting microscope.
  • TCM 199 with Earles salts (Gibco) 75.0 mg/l kanamycin, 30.0 mM Hepes, pH 7.4, osmolarity 280 mOsmols/Kg H 2 O
  • Matured oocytes were stripped of cumulus cells 18 hours after the onset of maturation. Denuded oocytes were then washed in calcium free M2 medium containing 10% Foetal Calf Serum (FCS) and maintained in this medium at 37° C. To remove the chromosomes (enucleation) oocytes were placed in calcium free M2 containing 10% FCS, 7.5 ug/ml cytochalasin B (Sigma) and 5.0 ug/ml Hoechst 33342 (Sigma) at 37° C. for 20 minutes. A small amount of cytoplasm from directly beneath the 1st polar body was then aspirated using a 20 ⁇ M glass pipette. Enucleation was confirmed by exposing the aspirated portion of cytoplasm to UV light and checking for the presence of a metaphase plate.
  • FCS Foetal Calf Serum
  • Enucleated oocytes were then used for each of the three methods of reconstruction (a), (b) and (c) as detailed below.
  • Enucleated oocytes were maintained in calcium free M2 10% FCS at 39° C. as soon as possible after enucleation, a single cell was placed into contact with the oocyte by using a glass pipette to transfer the cell through the hole previously made in the zona pellucida.
  • the cytoplast/cell couplet was then transferred into the fusion chamber in 200 ⁇ l of 0.3M mannitol in distilled water. The couplet, was manually aligned between the electrodes.
  • An AC pulse of 3V was applied for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80secs.
  • the couplets were then washed in calcium free M2, 10% FCS at 37° C. and incubated in the same medium under oil at 37° C.
  • Enucleated oocytes were returned to the maturation medium. At 30 or 42 hours post onset of maturation a single cell was placed into contact with the enucleated oocyte. The couplet was transferred to the fusion chamber (see below) in 200 ⁇ l of 0.3M mannitol, 0.1 mM MgSO 4 , 0.001 mM CaCl 2 in distilled water. Fusion and activation were induced by application of an AC pulse of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 ⁇ secs. Couplets were then washed in TC199 10% FCS and incubated at 37° C. 5% CO 2 for 15-20 hours (30 hpm group) or 4-8 hours (42 hpm group) [The abbreviation “hpm” is standard for “hours post-maturation”].
  • Enucleated oocytes were activated (as described below) 30 or 42 hours post onset of maturation and then cultured in TC199 10% FCS at 37° C. 5% CO 2 for 8-10 hours (30 hpm group) or 4-6 hours (42 hpm group). A single cell was then placed into contact with the oocyte and fusion induced as described below. The couplets were then cultured in TC199 10% FCS at 37° C. 5% CO 2 for a further 12-16 hours (30 hpm group) or 4-6 hours (42 hpm group).
  • oocytes were placed between two parallel electrodes in 200 ⁇ l of 0.3M mannitol, 0.1 mM MgSO 4 , 0.001 mM CaCl 2 in distilled water (Willadsen, Nature 320 63-65 (1986)). Activation was induced by application of 1 DC pulse of 1.25 kV/cm for 80 ⁇ s.
  • manipulated embryos were treated in a similar manner with the addition that the contact surface between the enucleated oocyte and the cell was arranged parallel to the electrodes. Fusion was induced by application of an AC current of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 ⁇ s.
  • couplets were double embedded in 1% and 1.2% agar (DIFCO) in PBS and transferred to the ligated oviduct of unsynchronised ewes (ewes are a less expensive alternative as a temporary recipient for the reconstructed embryo).
  • the couplet is embedded in agar to prevent or reduce immune rejection of the embryo by the recipient ewe and to assist in holding the couplet together.
  • recipient ewes were sacrificed and the embryos retrieved by flushing from the oviduct using PBS 10% FCS. Embryos were dissected from the agar chips using 2 needles and development assessed by microscopy.
  • In vitro techniques may also be suitable in place of a temporary recipient ewe to achieve development of the embryo to the blastocyst stage.
  • Table 2 shows the results of induction of pregnancy following transfer of all morula/blastocyst stage reconstructed embryos to the uterine horn of synchronised final recipient blackface ewes.
  • the Table shows the total number of embryos from each group transferred and the frequency of pregnancy in terms of ewes and embryos (in the majority of cases 2 embryos were transferred to each ewe.
  • a single twin pregnancy was established using the “MAGIC” cytoplast.
  • Table 3 shows the outcome of the pregnancies established following transfer or morula/blastocyst stage embryos to final recipient ewes.
  • EWE Method Passage Result 4E468 GOAT 6 LIVE LAMB 4E302 GOAT 7 FOETUS DIED (APPROX 130 DAYS) 4E210 GOAT 11 LIVE LAMB 4E286 GOAT 11 LIVE LAMB (DIED SHORTLY AFTER BIRTH) 4E453 GOAT 11 FOETUS DIED (APPROX 80 DAYS) 4E294 UNIVERSAL 11 LIVE LAMB 4E272 MAGIC 13 LIVE LAMB (DIED SHORTLY AFTER BIRTH)
  • OME ovine mammary epithelial
  • BLWF1 Black Welsh Fibroblast
  • SEC1 Sheep embryonic Cell
  • SEC1 cells are an epithelial-like cell line derived from a day 9 embryo obtained from a super ovulated and mated Pol-Dorset ewe to a Pol-Dorset tup.
  • the SEC1 cells are distinct from the TNT cells described in co-pending PCT application No. PCT/GB95/02095 published as WO 96/07732 for the following reasons. Firstly, the morphology of the cells of the two cell lines are completely different and secondly, the methods used to isolate the cell lines were different.
  • the SEC1 cell line was established from a single embryo whereas the TNT cell lines are derived from groups of cells.
  • Table 4 shows a summary of the development of nuclear transfer embryos reconstructed from different cell types. The table shows the number of embryos reconstructed, development to the blastocyst stage and number of pregnancies for each of the three cell types. All cell lines were karyotyped prior to their use for embryo reconstruction. These cell lines had a modal number of 54 chromosomes. One to three blastocyst stage embryos were transferred to each synchronised final recipient ewe. Reconstructed embryos which were cultured in vitro were placed into 10 ⁇ l (4 embryos) drops of SOFM (synthetic oviduct fluid medium) containing 10% human serum and cultured in a humidified atmosphere of 5% O 2 , 5% CO 2 and 90% N 2 at 39° C. Cultured embryos were transferred to fresh medium every two days. SOFs medium was prepared according to Gardner et al., Biology of Reproduction 50 390-400 (1994) and Thompson et al., Biology of Reproduction 53 1385-1391 (1995).
  • SOFM synthetic

Abstract

A method of reconstituting an animal embryo involves transferring the nucleus from a quiescent donor cell into a suitable recipient cell. The donor cell is quiescent, in that it is caused to exit from the growth and division cycle at G1 and to arrest in the G0 state. Nuclear transfer may take place by cell fusion. The reconstituted embryo may then give rise to one or more animals. The invention is useful in the production of transgenic animals as well as non-transgenics of high genetic merit.

Description

  • This invention relates to the generation of animals including but not being limited to genetically selected and/or modified animals.
  • The reconstruction of mammalian embryos by the transfer of a nucleus from a donor embryo to an enucleated oocyte or one cell zygote allows the production of genetically identical individuals. This has clear advantages for both research (i.e. as biological controls) and also in commercial applications (i.e. multiplication of genetically valuable livestock, uniformity of meat products, animal management). One problem with the use of early embryos as nuclear donors is that the number of offspring which can be produced from a single embryo is limited both by the number of cells (embryos at the 32-64 cell stage are the most widely used in farm animal species) and the efficiency of the nuclear transfer protocol.
  • In contrast to the use of embryos as nuclear donors, the ability to produce live offspring by nuclear transfer from cells which can be maintained in culture is an objective which have been sought for some time by animal breeders. The ability to produce cloned offspring from a cultured cell line would offer a large number of advantages over the use of early embryos. These include: the production of large numbers of identical offspring over a long time period (cultured cells can be frozen and stored) and the ability genetically to modify and/or select cell populations of the required genotype (e.g. sex) prior to embryo reconstruction. One potential cell type for use in these procedures is the Embryonic Stem (ES) cell. ES cells have been isolated in the mouse, however as yet there are no reports of development to term following their use in nuclear transfer. At the present time there is a single report of ES like cells in pig which have contributed to development following injection into the blastocoele cavity of in vivo-produced blastocysts (Wheeler, Reprod. Fertil. Dev. 6 563-568 (1994)) but no reports of chimerism in other farm livestock species and no reports of development to term following nuclear transfer in any mammalian species from any established cell line.
  • There are several alternatives to the use of ES cell lines; one of these is to search for other cell populations which are able to promote development when used for nuclear transfer. Several reports have suggested that Primordial Germ Cells offer a suitable candidate; however no development to term has yet been reported. Cell lines established from early embryos have been suggested; although development has been reported from early passage cells in the sheep (Campbell et al., Therio 43 181 (1995)) on prolonged culture, no development was obtained using conventional nuclear transfer protocols (Campbell et al., J. Abstract Series (5) 31 (1995)).
  • In order to obtain development to term after nuclear transfer the developmental clock of the transferred nucleus must be reset. For this to occur transcription must be arrested and then restarted in a developmentally regulated pattern. Previous reports have shown that development to the blastocyst stage can be obtained from a wide range of cell types in the cow, sheep, pig, rabbit and mouse. However, in all of these reports no development to term has been reported. The birth of live lambs following nuclear transfer from primary cell lines (up to and including passage 3) which were established from the embryonic disc (ED) of day 9 ovine embryos has previously been reported (Campbell et al., Therio 43 181 (1995)). However, on subsequent culture no development to term was obtained using conventional nuclear transfer protocols (at passage 6 and 11) (Campbell et al., J. Reprod. Fertil. Abstract Series (5) 31 (1995)). These results can, be interpreted in a number of ways; firstly it can be postulated that all of the ED derived cells obtained during early periods of culture are able to promote development. However, on prolonged culture during establishment of a cultured cell line these cells change and are thus unable to control development when used as nuclear donors for nuclear transfer into the “Universal Recipient” referred to in the above papers. Alternatively it may be postulated that during the early culture period a sub-population of cells retains the ability to promote development and that this would explain the production of live offspring following nuclear transfer during these early passages. Previous studies have emphasised the role of cell cycle co-ordination of the donor nucleus and the recipient cytoplasm in the development of embryos reconstructed by nuclear transfer (Campbell et al., Biol. Reprod. 49 933-942 (1993) and Biol. Reprod. 50 1385-1393 (1994)).
  • Two possible alternative strategies to that of relying on the isolation of a cell line which is totipotent for nuclear transfer using published nuclear transfer protocols are:
      • (1) to modify existing nuclear transfer procedures; or
      • (2) to modify the chromatin of the donor cell prior to nuclear transfer.
  • A totipotent cell can direct the development of a whole animal (when constructing embryos by nuclear transfer from a donor cell into a recipient cell, such as an enucleated oocyte, it is the nucleus of the donor cell which is totipotent). This includes directing the development of extra-embryonic lineages, i.e. the placenta. In this definition, a fertilised zygote and in some species individual blastomeres are also totipotent. In contradistinction, a pluripotent or multipotent cell (i.e. an embryonic stem cell) type has been defined as one which can form all tissues in the conceptus/offspring after injection into the blastocoele cavity.
  • In both the nuclear transfer strategies (1) and (2) outlined above, a method is required which will allow the reprogramming of gene expression of the transferred nucleus: such a method would then allow the use of differentiated or partially differentiated cells as nuclear donors and would “bring out” their inherent totipotency.
  • It has now been found that quiescent cells, that is to say cells which are not actively proliferating by means of the cell cycle, can advantageously be used as nuclear donors in the reconstitution of an animal embryo. Such embryos may then be allowed to develop to term. It seems that changes in the donor nucleus which are observed after embryo reconstruction and which are required for efficient nuclear transfer can be induced in the nuclei of cells prior to their use as nuclear donors by causing them to enter the quiescent state. This fact has been exploited in the present application.
  • According to a first aspect of the present invention, there is provided a method of reconstituting an animal embryo, the method comprising transferring the nucleus of a quiescent donor cell into a suitable recipient cell.
  • In principle, the invention is applicable to all animals, including birds, such as domestic fowl, amphibian species and fish species. In practice, however, it will be to non-human animals, especially (non-human) mammals, particularly placental mammals, that the greatest commercially useful applicability is presently envisaged. It is with ungulates, particularly economically important ungulates such as cattle, sheep, goats, water buffalo, camels and pigs that the invention is likely to be most useful, both as a means for cloning animals and as a means for generating transgenic or genetically modified animals. It should also be noted that the invention is also likely to be applicable to other economically important animal species such as, for example, horses, llamas or rodents e.g. rats or mice, or rabbits.
  • The invention is equally applicable in the production of transgenic, as well as non-transgenic animals. Transgenic animals may be produced from genetically altered donor cells. The overall procedure has a number of advantages over conventional procedures for the production of transgenic (i.e. genetically modified) animals which may be summarised as follows:
      • (1) fewer recipients will be required;
      • (2) multiple syngeneic founders may be generated using clonal donor cells;
      • (3) subtle genetic alteration by gene targeting is permitted;
      • (4) all animals produced from embryos prepared by the invention should transmit the relevant genetic modification through the germ line as each animal is derived from a single nucleus; in contrast, production of transgenic animals by pronuclear injection or chimerism after inclusion of modified stem cell populations by blastocyst injection, or other procedures, produces a proportion of mosaic animals in which all cells do not contain the modification and the resultant animal may not transmit the modification through the germ line; and
      • (5) cells can be selected for the site of genetic modification (e.g. integration) prior to the generation of the whole animal.
  • It should be noted that the term “transgenic”, in relation to animals, should not be taken to be limited to referring to animals containing in their germ line one or more genes from another species, although many transgenic animals will contain such a gene or genes. Rather, the term refers more broadly to any animal whose germ line has been the subject of technical intervention by recombinant DNA technology. So, for example, an animal in whose germ line an endogenous gene has been deleted, duplicated, activated or modified is a transgenic animal for the purposes of this invention as much as an animal to whose germ line an exogenous DNA sequence has been added.
  • In embodiments of the invention in which the animal is transgenic, the donor nucleus is genetically modified. The donor nucleus may contain one or more transgenes and the genetic modification may take place prior to nuclear transfer and embryo reconstitution. Although micro-injection, analogous to injection into the male or female pronucleus of a zygote, may be used as a method of genetic modification, the invention is not limited to that methodology: mass transformation or transfection techniques can also be used e.g. electroporation, viral transfection or lipofection.
  • In the method of the invention described above, a nucleus is transferred from a quiescent donor cell to a recipient cell. The use of this method is not restricted to a particular donor cell type. All cells of normal karyotype, including embryonic, foetal and adult somatic cells, which can be induced to enter quiescence or exist in a quiescent state in vivo may prove totipotent using this technology. The invention therefore contemplates the use of an at least partially differentiated cell, including a fully differentiated cell. Donor cells may be, but do not have to be, in culture. Cultured bovine primary fibroblasts, an embryo-derived ovine cell line (TNT4), an ovine mammary epithelial cell derived cell line (OME) from a 6 year old adult sheep, a fibroblast cell line derived from foetal ovine tissue (BLWF1) and an epithelial-like cell line derived from a 9-day old sheep embryo (SECL) are exemplified below. A class of embryo-derived cell lines useful in the invention which includes the TNT4 cell line are also the subject of co-pending PCT Patent Application No. PCT/GB95/02095, published as WO96/07732.
  • To be useful in the invention, donor cells are quiescent, which is to say that they are not actively proliferating by means of the mitotic cell cycle. The mitotic cell cycle has four distinct phases, G1, S, G2 and M. The beginning event in the cell cycle, called start, takes place in the G1 phase and has a unique function. The decision or commitment to undergo another cell cycle is made at start. Once a cell has passed through start, it passes through the remainder of the G1 phase, which is the pre-DNA synthesis phase. The second stage, the S phase, is when DNA synthesis takes place. This is followed by the G2 phase, which is the period between DNA synthesis and mitosis. Mitosis itself occurs at the M phase. Quiescent cells (which include cells in which quiescence has been induced as well as those cells which are naturally quiescent, such as certain fully differentiated cells) are generally regarded as not being in any of these four phases of the cycle; they are usually described as being in a G0 state, so as to indicate that they would not normally progress through the cycle. The nuclei of quiescent G0 cells have a diploid DNA content.
  • Cultured cells can be induced to enter the quiescent state by various methods including chemical treatments, nutrient deprivation, growth inhibition or manipulation of gene expression. Presently the reduction of serum levels in the culture medium has been used successfully to induce quiescence in both ovine and bovine cell lines. In this situation, the cells exit the growth cycle during the G1 phase and arrest, as explained above, in the so-called G0 stage. Such cells can remain in this state for several days (possibly longer depending upon the cell) until re-stimulated when they re-enter the growth cycle. Quiescent cells arrested in the G0 state are diploid. The G0 state is the point in the cell cycle from which cells are able to differentiate. On quiescence a number of metabolic changes have been reported and these include: monophosphorylated histones, ciliated centrioles, reduction or complete cessation in all protein synthesis, increased proteolysis, decrease in transcription and increased turnover of RNA resulting in a reduction in total cell RNA, disaggregation of polyribosomes, accumulation of inactive 80S ribosomes and chromatin condensation (reviewed Whitfield et al., Control of Animal Cell Proliferation, 1 331-365 (1985)).
  • Many of these features are those which are required to occur following transfer of a nucleus to an enucleated oocyte. The fact that the G0 state is associated with cell differentiation suggests that this may provide a nuclear/chromatin structure which is more amenable to either remodelling and/or reprogramming by the recipient cell cytoplasm. In this way, by virtue of the nuclear donor cells being in the quiescent state, the chromatin of the nuclei of the donors may be modified before embryo reconstitution or reconstruction such that the nuclei are able to direct development. This differs from all previously reported methods of nuclear transfer in that the chromatin of donor cells is modified prior to the use of the cells as nuclear donors.
  • The recipient cell to which the nucleus from the donor cell is transferred may be an oocyte or another suitable cell.
  • Recipient cells at a variety of different stages of development may be used, from oocytes at metaphase I through metaphase II, to zygotes and two-cell embryos. Each has its advantages and disadvantages. The use of fertilized eggs ensures efficient activation whereas parthenogenetic activation is required with oocytes (see below). Another mechanism that may favour the use of cleavage-stage embryos in some species is the extent to which reprogramming of gene expression is required. Transcription is initiated during the second cell cycle in the mouse and no major changes in the nature of the proteins being synthesised are revealed by two-dimensional electrophoresis until the blastocyst stage (Howlett & Bolton J. Embryol. Exp. Morphol. 87 175-206 (1985)). In most cases, though, the recipient cells will be oocytes.
  • It is preferred that the recipient be enucleate. While it has been generally assumed that enucleation of recipient oocytes in nuclear transfer procedures is essential, there is no published experimental confirmation of this judgement. The original procedure described for ungulates involved splitting the cell into two halves, one of which was likely to be enucleated (Willadsen Nature 320 (6) 63-65 (1986)). This procedure has the disadvantage that the other unknown half will still have the metaphase apparatus and that the reduction in volume of the cytoplasm is believed to accelerate the pattern of differentiation of the new embryo (Eviskov et al., Development 109 322-328 (1990)).
  • More recently, different procedures have been used in attempts to remove the chromosomes with a minimum of cytoplasm. Aspiration of the first polar body and neighbouring cytoplasm was found to remove the metaphase II apparatus in 67% of sheep oocytes (Smith & Wilmut Biol. Reprod. 40 1027-1035 (1989)). Only with the use of DNA-specific fluorochrome (Hoechst 33342) was a method provided by which enucleation would be guaranteed with the minimum reduction in cytoplasmic volume (Tsunoda et al., J. Reprod. Fertil. 82 173 (1988)). In livestock species, this is probably the method of routine use at present (Prather & First J. Reprod. Fertil. Suppl. 41 125 (1990), Westhusin et al., Biol. Reprod. (Suppl.) 42 176 (1990)).
  • There have been very few reports of non-invasive approaches to enucleation in mammals, whereas in amphibians, irradiation with ultraviolet light is used as a routine procedure (Gurdon Q. J. Microac. Soc. 101 299-311 (1960)). There are no detailed reports of the use of this approach in mammals, although during the use of DNA-specific fluorochrome it was noted that exposure of mouse oocytes to ultraviolet light for more than 30 seconds reduced the developmental potential of the cell (Tsunoda et al., J. Reprod. Fertil. 82 173 (1988)).
  • It is preferred that recipient host cells to which the donor cell nucleus is transferred is an enucleated metaphase II oocyte, an enucleated unactivated oocyte or an enucleated preactivated oocyte. At least where the recipient is an enucleated metaphase II oocyte, activation may take place at the time of transfer. Alternatively, at least where the recipient is an enucleated unactivated metaphase II oocyte, activation may take place subsequently. As described above enucleation may be achieved physically, by actual removal of the nucleus, pro-nuclei or metaphase plate (depending on the recipient cell), or functionally, such as by the application of ultraviolet radiation or another enucleating influence.
  • Three suitable cytoplast (enucleated oocyte) recipients are:
  • 1. The “MAGIC Recipient” (Metaphase Arrested G1/G0AcceptIng Cytoplast) described in our co-pending PCT patent application No. PCT/GB96/02098 filed today (claiming priority from GB 9517779.6).
  • 2. The “GOAT” (G0/G1 Activation and Transfer)—a MII (metaphase II) oocyte at the time of activation (Campbell et al., Biol. Reprod. 49 933-942 (1993).
  • 3. The “Universal Recipient” (Campbell et al., Biol. Reprod. 649 933-942 (1993), Biol. Reprod. 50 1385-1393 (1994).
  • All three of these recipients would result in normal ploidy when using donor nuclei in Go in the reconstructed embryo. However, recent reports have suggested that a proportion of the nuclei from quiescent cells are unable to enter the DNA synthetic phase when placed into an S-phase cytoplasm without undergoing disassembly of the nuclear envelope (Leno & Munshi, J. Cell Biol. 127(1) 5-14 (1994)). Therefore, although a proportion of embryos will develop when using the “Universal Recipient” it is postulated that the use of MII oocytes containing high levels of MPF (M-phase promoting factor or maturation-promoting factor) as cytoplast recipients by either method 1 or 2 will result in a greater frequency of development.
  • Once suitable donor and recipient cells have been identified, it is necessary for the nucleus of the former to be transferred to the latter. Most conveniently, nuclear transfer is effected by fusion.
  • Three established methods which have been used to induce fusion are:
      • (1) exposure of cells to fusion-promoting chemicals, such as polyethylene glycol;
      • (2) the use of inactivated virus, such as Sendai virus; and
      • (3) the use of electrical stimulation.
  • Exposure of cells to fusion-promoting chemicals such as polyethylene glycol or other glycols is a routine procedure for the fusion of somatic cells, but it has not been widely used with embryos. As polyethylene glycol is toxic it is necessary to expose the cells for a minimum period and the need to be able to remove the chemical quickly may necessitate the removal of the zona pellucida (Kanka et al., Mol. Reprod. Dev. 29 110-116 (1991)). In experiments with mouse embryos, inactivated Sendai virus provides an efficient means for the fusion of cells from cleavage-stage embryos (Graham Wistar Inst. Symp. Monogr. 9 19 (1969)), with the additional experimental advantage that activation is not induced. In ungulates, fusion is commonly achieved by the same electrical stimulation that is used to induce parthogenetic activation (Willadsen Nature 320 (6) 63-65 (1986), Prather et al., Biol. Reprod. 37 859-866 (1987)). In these species, Sendai virus induces fusion in a proportion of cases, but is not sufficiently reliable for routine application (Willadsen Nature 320 (6) 63-65 (1986)).
  • While cell-cell fusion is a preferred method of effecting nuclear transfer, it is not the only method that can be used. Other suitable techniques include microinjection (Ritchie and Campbell, J. Reproduction and Fertility Abstract Series No. 15, p60).
  • Before or (preferably) after nuclear transfer (or, in some instances at least, concomitantly with it), it is generally necessary to stimulate the recipient cell into development by parthenogenetic activation, at least if the cell is an oocyte. Recent experiments have shown that the requirements for parthogenetic activation are more complicated than had been imagined. It had been assumed that activation is an all-or-none phenomenon and that the large number of treatments able to induce formation of a pronucleus were all causing “activation”. However, exposure of rabbit oocytes to repeated electrical pulses revealed that only selection of an appropriate series of pulses and control of the Ca2+ was able to promote development of diploidized oocytes to mid-gestation (Ozil Development 109 117-127 (1990)). During fertilization there are repeated, transient increases in intracellular calcium concentration (Cutbertson & Cobbold Nature 316 541-542 (1985)) and electrical pulses are believed to cause analogous increases in calcium concentration. There is evidence that the pattern of calcium transients varies with species and it can be anticipated that the optimal pattern of electrical pulses will vary in a similar manner. The interval between pulses in the rabbit is approximately 4 minutes (Ozil Development 109 117-127 (1990)), and in the mouse 10 to 20 minutes (Cutbertson & Cobbold Nature 316 541-542 (1985)), while there are preliminary observations in the cow that the interval is approximately 20 to 30 minutes (Robl et al., in Symposium on cloning Mammals by Nuclear Transplantation (Seidel ed.), Colorado State University, 24-27 (1992)). In most published experiments activation was induced with a single electrical pulse, but new observations suggest that the proportion of reconstituted embryos that develop is increased by exposure to several pulses (Collas & Robl Biol. Reprod. 43 877-884 (1990)). In any individual case, routine adjustments may be made to optimise the number of pulses, the field strength and duration of the pulses and the calcium concentration of the medium.
  • According to a second aspect of the present invention there is provided a reconstituted animal embryo prepared by a method as described previously.
  • According to a third aspect of the present invention there is provided a method for preparing an animal, the method comprising:
      • (a) reconstituting an animal embryo as described above; and
      • (b) causing an animal to develop to term from the embryo; and
      • (c) optionally, breeding from the animal so formed.
  • Step (a) has been described in depth above.
  • The second step, step (b) in the method of this aspect of the invention is to cause an animal to develop to term from the embryo. This may be done directly or indirectly. In direct development, the reconstituted embryo from step (a) is simply allowed to develop without further intervention beyond any that may be necessary to allow the development to take place. In indirect development, however, the embryo may be further manipulated before full development takes place. For example, the embryo may be split and the cells clonally expanded, for the purpose of improving yield.
  • Alternatively or additionally, it may be possible for increased yields of viable embryos to be achieved by means of the present invention by clonal expansion of donors and/or if use is made of the process of serial (nuclear) transfer. A limitation in the presently achieved rate of blastocyst formation may be due to the fact that a majority of the embryos do not “reprogram” (although an acceptable number do). If this is the case, then the rate may be enhanced as follows. Each embryo that does develop itself can be used as a nuclear donor, such as, for example at the morula or 32-64 cell stage; alternatively, inner cell mass cells can be used at the blastocyst stage. Embryos derived from these subsequent transfers could themselves also be used as potential nuclear donors to further increase efficiency. If these embryos do indeed reflect those which have reprogrammed gene expression and those nuclei are in fact reprogrammed (as seems likely), then each developing embryo may be multiplied in this way by the efficiency of the nuclear transfer process. The degree of enhancement likely to be achieved depends upon the cell type. In sheep, it is readily possible to obtain 55% blastocyst stage embryos by transfer of a single blastomere from a 16 cell embryo to a preactivated “Universal Recipient” oocyte. So it is reasonable to hypothesise that each embryo developed from a single cell could give rise to eight at the 16 cell stage. Although these figures are just a rough guide, it is clear that at later developmental stages the extent of benefit would depend on the efficiency of the process at that stage.
  • It is also contemplated that a new cell line to act as a source of nuclear donor cells could be produced from embryos formed according to the preceding description or the resulting foetuses or adults.
  • In certain instances, where there may be restrictions in the development of a reconstructed embryo to term it may be preferable to generate a chimeric animal formed from cells derived from a naturally formed embryo and an embryo reconstructed by nuclear transfer. Such a chimera can be formed by taking a proportion of cells of the natural embryo and a proportion of the cells of the reconstructed embryo at any stage up to the blastocyst stage and forming a new embryo by aggregation or injection. The proportion of cells may be in the ratio of 50:50 or another suitable ratio to achieve the formation of an embryo which develops to term. The presence of normal cells in these circumstances is thought to assist in rescuing the reconstructed embryo and allowing successful development to term and a live birth.
  • Aside from the issue of yield-improving expediencies, the reconstituted embryo may be cultured, in viva or in vitro to blastocyst.
  • Experience suggests that embryos derived by nuclear transfer are different from normal embryos and sometimes benefit from or even require culture conditions in viva other than those in which embryos are usually cultured (at least in viva). The reason for this is not known. In routine multiplication of bovine embryos, reconstituted embryos (many of them at once) have been cultured in sheep oviducts for 5 to 6 days (as described by Willadsen, In Mammalian Egg Transfer (Adams, E. E., ed.) 185 CRC Press, Boca Raton, Fla. (1982)). In the practice of the present invention, though, in order to protect the embryo it should preferably be embedded in a protective medium such as agar before transfer and then dissected from the agar after recovery from the temporary recipient. The function of the protective agar or other medium is twofold: first, it acts as a structural aid for the embryo by holding the zona pellucida together; and secondly it acts as barrier to cells of the recipient animal's immune system. Although this approach increases the proportion of embryos that form blastocysts, there is the disadvantage that a number of embryos may be lost.
  • If in vitro conditions are used, those routinely employed in the art are quite acceptable.
  • At the blastocyst stage, the embryo may be screened for suitability for development to term. Typically, this will be done where the embryo is transgenic and screening and selection for stable integrants has been carried out. Screening for non-transgenic genetic markers may also be carried out at this stage. However, because the method of the invention allows for screening of donors at an earlier stage, that will generally be preferred.
  • After screening, if screening has taken place, the blastocyst embryo is allowed to develop to term. This will generally be in vivo. If development up to blastocyst has taken place in vitro, then transfer into the final recipient animal takes place at this stage. If blastocyst development has taken place in vivo, although in principle the blastocyst can be allowed to develop to term in the pre-blastocyst host, in practice the blastocyst will usually be removed from the (temporary). pre-blastocyst recipient and, after dissection from the protective medium, will be transferred to the (permanent) post-blastocyst recipient.
  • In optional step (c) of this aspect of the invention, animals may be bred from the animal prepared by the preceding steps. In this way, an animal may be used to establish a herd or flock of animals having the desired genetic characteristic(s).
  • Animals produced by transfer of nuclei from a source of genetically identical cells share the same nucleus, but are not strictly identical as they are derived from different oocytes. The significance of this different origin is not clear, but may affect commercial traits. Recent analyses of the mitochondrial DNA of dairy cattle in the Iowa State University Breeding Herd revealed associated with milk and reproductive performance (Freeman & Beitz, In Symposium on Cloning Mammals by Nuclear Transplantation (Seidel, G. E. Jr., ed.) 17-20, Colorado State University, Colorado (1992)). It remains to be confirmed that similar effects are present throughout the cattle population and to consider whether it is possible or necessary in specific situations to consider the selection of oocytes. In the area of cattle breeding the ability to produce large numbers of embryos from donors of high genetic merit may have considerable potential value in disseminating genetic improvement through the national herd. The scale of application will depend upon the cost of each embryo and the proportion of transferred embryos able to develop to term.
  • By way of illustration and summary, the following scheme sets out a typical process by which transgenic and non-transgenic animals may be prepared. The process can be regarded as involving seven steps:
      • (1) selection and isolation of suitable donor cells, which may include assessment of karyotype, induction of quiescence (arrest in G0) and/or induction of development;
      • (2) application of suitable molecular biological techniques for the production of genetically modified cell populations. Such techniques may include gene additions, gene knock-outs, gene knock-ins, and other gene modifications. Optionally, transgenesis, may also be employed by transfection with suitable constructs, with or without selectable markers;
      • (3) optionally screen and select modified cell populations or clones for the required is genotype/phenotype (i.e. stable integrants);
      • (4) induction of quiescence in modified cell population;
      • (5) embryo reconstitution by nuclear transfer;.
      • (6) culture, in vivo or in vitro, to blastocyst;
        • (6a) optionally screen and select for stable integrants—omit if done at (3)—or other desired characteristics;
      • (7) transfer if necessary to final recipient.
  • According to a fourth aspect of the invention, there is provided an animal prepared as described above.
  • Preferred features for each aspect of the invention are as for each other aspect, mutatis mutandis.
  • The present invention will now be described by reference to the accompanying Examples which are provided for the purposes of illustration and are not to be construed as being limiting on the present invention.
  • EXAMPLES Example 1 Induction of Quiescence in Donor Cells
  • Various methods have been shown to induce quiescence in cultured cell lines, including; contact inhibition or serum starvation (reviewed Whitfield et al., Control of Animal Cell Proliferation, 1 331-365 (1985)). The method of: induction of quiescence is not thought to be of importance, the important step is that the cells exit the growth cycle, arrest in a G0 state with a diploid DNA content and remain viable. In Examples 3 and 4, serum starvation of bovine primary fibroblasts, a bovine cell line established from the inner cell mass of day 7 in vivo produced blastocysts, and an embryo derived ovine cell line (TNT4), was used to induce quiescence and arrest the cells in the G0 phase of the cell cycle. Serum starvation was similarly used to induce quiescence of the donor cells described in Example 5.
  • Example 2 Isolation of Oocytes and Nuclear Transfer
  • Oocytes can be obtained by (i) in vitro maturation of slaughterhouse material, or from transvaginal follicle puncture; (ii) in vivo maturation and surgically recovery; or (iii) any other suitable procedure. All in vivo matured oocytes should be harvested by flushing from the oviduct in calcium magnesium free phosphate buffered saline (PBS) containing 1.0% foetal calf serum (FCS). In vitro matured oocytes are harvested and transferred to calcium free M2 (Whittingham and Wales Aust. J. Biol. Sci. 22 1065-1068 (1969)) containing 1.0% FCS. Oocytes are denuded of cumulus, cells and enucleated as previously described (Campbell et al., Biol. Reprod. 49 933-942 (1993) and Biol. Reprod. 50 1385-1393 (1994)) with the exception that calcium free medium is used for all procedures. Fusion procedures are modifications of those previously reported (Campbell et al., 1993, 1994 loc cit) and are as described in the relevant section below, alternatively the nucleus may be introduced by injection of the donor cell into the enucleated oocyte (Ritchie & Campbell, J. Reprod. Fertil. Abstract Series (5) 60 (1995)). The timing of these events is dependent upon the species, the following two protocols outline the use of in vivo matured ovine and in vitro matured bovine oocytes.
  • Example 3 Ovine Nuclear Transfer
  • 3.1 Superstimulation of Donor Ewes and Recovery of Oocytes
  • Scottish Blackface ewes were synchronised with progestagen sponges for 14 days (Veramix™, Upjohn, UK) and induced to superovulate with single injections of 3.0 mg/day (total 6.0 mg) ovine follicle-stimulating hormone (FSH) (Ovagen™, Immuno-chemical Products Ltd, New Zealand) on two successive days. Ovulation was induced with an 8 mg single dose of a gonadotropin-releasing hormone analogue (GnRH Receptal™, Hoechst, UK) 24 hours after the second injection of FSH.
  • Unfertilised metaphase II oocytes were recovered by flushing from the oviduct at 24-29 hours after GnRH injection using Dulbeccols phosphate buffered saline containing 1.0% foetal calf serum (FCS) maintained at 37° C. until use.
  • 3.2 Oocyte Manipulation
  • Recovered oocytes were maintained at 37° C., washed in PBS 1.0% FCS and transferred to calcium free M2 medium containing 10% Foetal Calf Serum (FCS), at 37° C. To remove the chromosomes, (enucleation) oocytes were placed in calcium free M2 containing 10% FCS, 7.5 μg/ml cytochalasin B (Sigma) and 5.0 μg/ml Hoechst 33342 (Sigma) at 37° C. for 20 minutes. A small amount of cytoplasm from directly beneath the 1st polar body was then aspirated using a 20 μM glass pipette. Enucleation was confirmed by exposing the aspirated portion of cytoplasm to UV light and checking for the presence of a metaphase plate.
  • 3.3 Embryo Reconstruction
  • Groups of 10-20 oocytes were enucleated and placed into 20 μl drops of calcium free M2 medium at 37° C. 5% CO2 under mineral oil (SIGMA). Each of the following three protocols (a), (b) and (c) were used for embryo reconstruction.
  • (a) “MAGIC” (Metaphase Arrested G1/G0 Accepting Cytoplast)
  • As soon as possible after enucleation a single cell was placed into contact with the oocyte by using a glass pipette to transfer the cell through the hole previously made in the zona pellucida. The cytoplast/cell couplet was then transferred into the fusion chamber in 200 μl of 0.3M mannitol in distilled water and manually aligned between the electrodes. An AC pulse of 5V was applied for 3 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 μsecs. The couplets were then washed in calcium free M2, 10% FCS at 37° C. and incubated in the same medium under oil at 37° C. 5% CO2. 30 minutes prior to activation the couplets were transferred to calcium free M2 medium 10% FCS containing 5 μM nocodazole. Activation was induced at 32-34 hours post hCG injection as described below. Following activation the reconstructed zygotes were incubated in medium TC199 (Gibco) 10% FCS at 37° C. 5% CO2 for a further 3 hours. They were then washed 3 times for 5 minutes at 37° C. in the same medium without nocodazole and cultured for a further 12-15 hours prior to transfer to temporary recipient ewes.
  • (b) “GOAT” (G0/G1 Activation and Transfer)
  • At 32-34 hours post hCG injection a single cell was placed into contact with the enucleated oocyte. The couplet was transferred to the fusion chamber (see below) in 200 μl of 0.3M mannitol, 0.1 mM MgSO4, 0.001 mM CaCl2 in distilled water. Fusion and activation were induced by application of an AC pulse of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 μsecs. Couplets were then washed in TC199 10% FCS containing 7.5 μg/ml cytochalasin B and incubated in this medium for 1 hour at 37° C. 5% CO2. Couplets were then washed in TC199 10% FCS and cultured for a further 12-15 hours in TC199 10% FCS at 37° C. 5% CO2.
  • (c) “Universal Recipient”
  • Enucleated oocytes were activated (as described below) 32-34 hours post hCG injection and then cultured in TC199 10% FCS at 37° C. 5% CO2 for 4-6 hours. A single cell was then placed into contact with the oocyte and fusion induced as described below. The couplets were then incubated in TC199 10% FCS 7.5 kg cytochalasin B for 1 hour at 37° C. 5% CO2. Couplets were then washed and cultured in TC199 10% FCS at 37° C. 5% CO2 for a further 8-11 hours.
  • 3.4 Fusion and Activation
  • For activation, oocytes were placed between two parallel electrodes in 200 μl of 0.3M mannitol, 0.1 mM MgSO4. 0.001 mM CaCl2 in distilled water (Willadsen, Nature 320 63-65 (1986)). Activation was induced by application of 1 DC pulse of 1.25 kV/cm for 80 μs. For fusion, manipulated embryos were treated in a similar manner with the addition that the contact surface between the enucleated oocyte and the cell was arranged parallel to the electrodes. Fusion was induced by application of an AC current of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 μs.
  • 3.5 Embryo Culture and Assessment (All Groups)
  • After the culture period fused couplets were double embedded in 1% and 1.2% agar (DIFCO) in PBS and transferred to the ligated oviduct of unsynchronised ewes. The couplet is embedded in agar to prevent or reduce immune rejection of the embryo by the recipient ewe and to assist in holding the couplet together. After 6 days recipient ewes were sacrificed and the embryos retrieved by flushing from the oviduct using PBS 10% FCS. Embryos were dissected from the agar chips using 2 needles and development assessed by microscopy. All embryos which had developed to the morula/blastocyst stage were transferred as soon as possible to the uterine horn of synchronised final recipient ewes.
  • In vitro techniques may also be suitable in place of a temporary recipient ewe to achieve development of the embryo to the blastocyst stage.
  • Example 4 Bovine Nuclear Transfer
  • 4.1 In Vitro Oocyte Maturation
  • Ovaries were obtained from a local abattoir and maintained at 28-32° C. during transport to the laboratory. Cumulus oocyte complexes (COC's) were aspirated from follicles 3-10 mm in diameter using a hypodermic needle (1.2 mm internal diameter) and placed into sterile plastic universal containers. The universal containers were placed into a warmed chamber (35° C.) and the follicular material allowed to settle for 10-15 minutes before pouring off three quarters of the supernatant. The remaining follicular material was diluted with an equal volume of dissection medium (TCM 199 with Earles salts (Gibco), 75.0 mg/l kanamycin, 30.0 mM Hepes, pH 7.4, osmolarity 280 mOsmols/Kg H2O) supplemented with 10% bovine serum, transferred into an 85 mm petri dish and searched for COC's under a dissecting microscope. Complexes with at least 2-3 compact layers of, cumulus cells were selected washed three times in dissection medium and transferred into maturation medium (TC medium 199 with Earles salts (Gibco), 75 mg/l kanamycin, 30.0 mM Hepes, 7.69 mM NaHCO3, pH 7.8, osmolarity 280 mOsmols/Kg H2O) supplemented with 10% bovine serum and 1×106 granulosa cells/ml and cultured until required on a rocking table at 39° C. in an atmosphere of 5% CO2 in air.
  • 4.2 Oocyte Manipulation
  • Matured oocytes were stripped of cumulus cells 18 hours after the onset of maturation. Denuded oocytes were then washed in calcium free M2 medium containing 10% Foetal Calf Serum (FCS) and maintained in this medium at 37° C. To remove the chromosomes (enucleation) oocytes were placed in calcium free M2 containing 10% FCS, 7.5 ug/ml cytochalasin B (Sigma) and 5.0 ug/ml Hoechst 33342 (Sigma) at 37° C. for 20 minutes. A small amount of cytoplasm from directly beneath the 1st polar body was then aspirated using a 20 μM glass pipette. Enucleation was confirmed by exposing the aspirated portion of cytoplasm to UV light and checking for the presence of a metaphase plate.
  • 4.3 Embryo Reconstruction
  • Enucleated oocytes were then used for each of the three methods of reconstruction (a), (b) and (c) as detailed below.
  • (a) “MAGIC” (Metaphase Arrested G1/G0 Accenting Cytoplast)
  • Enucleated oocytes were maintained in calcium free M2 10% FCS at 39° C. as soon as possible after enucleation, a single cell was placed into contact with the oocyte by using a glass pipette to transfer the cell through the hole previously made in the zona pellucida. The cytoplast/cell couplet was then transferred into the fusion chamber in 200 μl of 0.3M mannitol in distilled water. The couplet, was manually aligned between the electrodes. An AC pulse of 3V was applied for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80secs. The couplets were then washed in calcium free M2, 10% FCS at 37° C. and incubated in the same medium under oil at 37° C. 5% CO2. 30 minutes prior to activation the couplets were transferred to calcium free M2 medium 10% FCS containing 5 μM nocodazole. Activation was induced as described below, following activation the reconstructed zygotes were incubated in medium TC199 10% FCS at 37° C. 5% CO2 for a further 3 hours. They were then washed 3 times for 5 minutes at 37° C. in the same medium without nocodazole and cultured for a further 12-15 hours prior to transfer to temporary recipient ewes (ewes are a less expensive alternative as a temporary recipient for the reconstructed embryo).
  • (b) “GOAT” (G0/G1 Activation and Transfer)
  • Enucleated oocytes were returned to the maturation medium. At 30 or 42 hours post onset of maturation a single cell was placed into contact with the enucleated oocyte. The couplet was transferred to the fusion chamber (see below) in 200 μl of 0.3M mannitol, 0.1 mM MgSO4, 0.001 mM CaCl2 in distilled water. Fusion and activation were induced by application of an AC pulse of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 μsecs. Couplets were then washed in TC199 10% FCS and incubated at 37° C. 5% CO2 for 15-20 hours (30 hpm group) or 4-8 hours (42 hpm group) [The abbreviation “hpm” is standard for “hours post-maturation”].
  • (c) “Universal Recipient”
  • Enucleated oocytes were activated (as described below) 30 or 42 hours post onset of maturation and then cultured in TC199 10% FCS at 37° C. 5% CO2 for 8-10 hours (30 hpm group) or 4-6 hours (42 hpm group). A single cell was then placed into contact with the oocyte and fusion induced as described below. The couplets were then cultured in TC199 10% FCS at 37° C. 5% CO2 for a further 12-16 hours (30 hpm group) or 4-6 hours (42 hpm group).
  • 4.4 Fusion and Activation
  • For activation, oocytes were placed between two parallel electrodes in 200 μl of 0.3M mannitol, 0.1 mM MgSO4, 0.001 mM CaCl2 in distilled water (Willadsen, Nature 320 63-65 (1986)). Activation was induced by application of 1 DC pulse of 1.25 kV/cm for 80 μs. For fusion, manipulated embryos were treated in a similar manner with the addition that the contact surface between the enucleated oocyte and the cell was arranged parallel to the electrodes. Fusion was induced by application of an AC current of 3V for 5 seconds followed by 3 DC pulses of 1.25 kV/cm for 80 μs.
  • 4.5 Embryo Culture and Assessment (All Groups)
  • After the culture period fused couplets were double embedded in 1% and 1.2% agar (DIFCO) in PBS and transferred to the ligated oviduct of unsynchronised ewes (ewes are a less expensive alternative as a temporary recipient for the reconstructed embryo). The couplet is embedded in agar to prevent or reduce immune rejection of the embryo by the recipient ewe and to assist in holding the couplet together. After 6 days recipient ewes were sacrificed and the embryos retrieved by flushing from the oviduct using PBS 10% FCS. Embryos were dissected from the agar chips using 2 needles and development assessed by microscopy.
  • In vitro techniques may also be suitable in place of a temporary recipient ewe to achieve development of the embryo to the blastocyst stage.
  • Results of Example 3 (Ovine Cells) and Example 4 (Bovine Cells
  • The present techniques have been applied to both ovine and bovine embryo reconstruction. At the present time blastocyst stage embryos have been obtained in cattle; however, no transfers of these embryos to final recipients have been performed. In sheep 7 recipient ewes became pregnant resulting in the birth of 5 live lambs (2 of which died shortly after birth). The results from these experiments are summarised in Tables 1-3.
  • Table 1 shows the results of development to blastocyst stage of ovine embryos reconstructed using quiescent TNT4 cell populations and 3 different cytoplast recipients. Reconstructed embryos were cultured in the ligated oviduct of a temporary recipient ewe until Day 7 after reconstruction. The results are expressed as the percentage of morula/blastocyst stage embryos in relation to the total number of embryos recovered.
    TABLE 1
    NUMBER MORULAE,
    BLASTOCYSTS/TOTAL
    DATE OF PAS- NUMBER OF
    NUCLEAR SAGE COUPLETS RECOVERED
    TRANSFER NUMBER “GOAT” “MAGIC” “UNIVERSAL”
    17.1.95 6  6/32  4/28
    19.1.95 7  1/26  1/10
    31.1.95 13  0/2 2/14
     2.2.95 13  0/11  0/14
     7.2.95 11  1/9 0/9
     9.2.95 11  9/29  1/2
    14.2.95 12 6/45
    16.2.95 13  3/13
    TOTAL 16/98 (16.3%) 10/78 8/68 (11.7%)
    (12.8%)
  • Table 2 shows the results of induction of pregnancy following transfer of all morula/blastocyst stage reconstructed embryos to the uterine horn of synchronised final recipient blackface ewes. The Table shows the total number of embryos from each group transferred and the frequency of pregnancy in terms of ewes and embryos (in the majority of cases 2 embryos were transferred to each ewe. A single twin pregnancy was established using the “MAGIC” cytoplast.
    TABLE 2
    PASSAGE
    NUMBER “MAGIC” “GOAT” “UNIVERSAL”
    P6 4 6 0
    P7 1 1 0
    P11 2 9 0
    P12 0 0 6
    P13 3 0 2
    TOTAL MOR/BL 10 16 8
    TOTAL NUMBER 6 9 4
    EWES
    PREGNANT EWES %   1 (16.7)   5 (55.5)   1 (25.0)
    FOETUSES/ 2/10 (20.0) 5/16 (31.25) 1/8 (12.5)
    TOTAL
    TRANSFERRED
    (%)
  • Table 3 shows the outcome of the pregnancies established following transfer or morula/blastocyst stage embryos to final recipient ewes.
    TABLE 3
    EWE Method Passage Result
    4E468 GOAT 6 LIVE LAMB
    4E302 GOAT 7 FOETUS DIED
    (APPROX 130 DAYS)
    4E210 GOAT 11 LIVE LAMB
    4E286 GOAT 11 LIVE LAMB (DIED
    SHORTLY AFTER BIRTH)
    4E453 GOAT 11 FOETUS DIED
    (APPROX 80 DAYS)
    4E294 UNIVERSAL 11 LIVE LAMB
    4E272 MAGIC 13 LIVE LAMB (DIED
    SHORTLY AFTER BIRTH)
  • Example 5 Ovine Nuclear Transfer and Embryo Reconstruction Using OME, BLWF1 and SEC1 Cells
  • Nuclear transfer has been conducted using three new cell types, designated OME, BLWF1 and SEC1. OME (ovine mammary epithelial) cells are an epithelial cell line established from a biopsy removed from the mammary gland of an adult 6 year old Fin-Dorset ewe, following the procedure of Finch et al., (Biochem. Soc. Trans. 24 369S (1996). BLWF1 (Black Welsh Fibroblast) cells are a fibroblast cell line obtained by dissection and culture of a day 26 Black Welsh foetus obtained following Natural Mating of a Black Welsh ewe to a Black Welsh tup. The method of isolation of primary foetal fibroblasts is according to Robertson, E. J., in Teratocarcinomas and embryonic stem cells: A practical approach, 71-112, IRL Press Oxford (1987). SEC1 (Sheep embryonic Cell) are an epithelial-like cell line derived from a day 9 embryo obtained from a super ovulated and mated Pol-Dorset ewe to a Pol-Dorset tup. The SEC1 cells are distinct from the TNT cells described in co-pending PCT application No. PCT/GB95/02095 published as WO 96/07732 for the following reasons. Firstly, the morphology of the cells of the two cell lines are completely different and secondly, the methods used to isolate the cell lines were different. The SEC1 cell line was established from a single embryo whereas the TNT cell lines are derived from groups of cells.
  • All cell lines were karyotyped and showed a modal chromosome number of 54 (2n). Prior to use as nuclear donors for embryo reconstruction, the induction of quiescence following the reduction of serum levels to 0.5% was monitored as previously described (Campbell et al., Nature 380 64-66 (1996)). Preparation of the reconstructed embryos was as described above in the previous examples.
  • Table 4 shows a summary of the development of nuclear transfer embryos reconstructed from different cell types. The table shows the number of embryos reconstructed, development to the blastocyst stage and number of pregnancies for each of the three cell types. All cell lines were karyotyped prior to their use for embryo reconstruction. These cell lines had a modal number of 54 chromosomes. One to three blastocyst stage embryos were transferred to each synchronised final recipient ewe. Reconstructed embryos which were cultured in vitro were placed into 10 μl (4 embryos) drops of SOFM (synthetic oviduct fluid medium) containing 10% human serum and cultured in a humidified atmosphere of 5% O2, 5% CO2 and 90% N2 at 39° C. Cultured embryos were transferred to fresh medium every two days. SOFs medium was prepared according to Gardner et al., Biology of Reproduction 50 390-400 (1994) and Thompson et al., Biology of Reproduction 53 1385-1391 (1995).
  • Table 5 shows the identification of the recipient ewes remaining pregnant at 24 Jun. 1996, the cell type used for embryo reconstruction and the expected lambing date. Pregnancies were established by the transfer of 1 to 3 morula/blastocyst stage embryos (on day 7 after reconstruction) to synchronised final recipient ewes. Details of the numbers reconstructed are shown in Table 4. Abbreviations are: PD=Pol-Dorset, BW=Black Welsh, FD=Fin-Dorset, *=embryo cultured in vitro to the blastocyst stage.
    TABLE 4
    No.
    couplets
    No. couplets recovered
    No. of No. fused transferred to from No. Morula/ No. pregnancies/
    Cell cytoplasts couplets oviducts (in oviduct Blastocyst No. pregnancies/ No. recipient ewes
    Type prepared (%) vitro cultured) (%) stage (%) No. Blastocyst (%) (%)
    OME 387 277 (63.8) 277 247 (89.2) 29 (11.7)  1/29 (3.4)  1/13 (7.7)
    BLWF1 203 172 (84.7) 143 (24) 124 (86.7) 34 (27.4)  4/34 (11.8)  4/10 (40.0)
    13 (54.2)  1/6 (16.6)  1/6 (16.6)
    SEC1 465 385 (82.8) 271 (92) 231 (85.3) 90 (39.0) 14/72 (19.5) 14/27 (51.8)
    36 (39.0)  1/15 (6.6)  1/5 (20.0)
  • TABLE 5
    I.D. of recipient ewe Cell line Outcome of pregnancy Breed
    5E191 SEC1 LIVE LAMB (MALE) PD
    5E17 SEC1 LIVE LAMB (MALE) PD
    5E134 SEC1 DEAD LAMB (MALE) PD
    9M399 SEC1 LIVE LAMB (MALE) PD
    5E524 SEC1 LIVE LAMB (MALE) PD
    5E139 BLWF1 LIVE LAMB (MALE) BW
    5E328 BLWF1 LIVE LAMB (MALE) BW
    5E169 BLWF1 LIVE LAMB (MALE) BW
    died at birth
    5E475 OME LIVE LAMB (FEMALE) FD

Claims (9)

1-18. (canceled)
19. A reconstructed ungulate embryo comprising a nucleus from a quiescent, ungulate somatic cell;
wherein the embryo is capable of developing to term.
20. The embryo of claim 19, wherein the ungulate is a bovine.
21. The embryo of claim 19, wherein the ungulate is a sheep.
22. The embryo of claim 19, wherein the ungulate is a goat.
23. The embryo of claim 19, wherein the ungulate is a pig.
24. The embryo of claim 19, wherein the somatic cell is a fibroblast cell.
25. The embryo of claim 19, wherein the somatic cell is a cultured cell.
26. The embryo of claim 19, wherein the somatic cell is a transgenic cell.
US11/543,786 1995-08-31 2006-10-06 Quiescent cell populations for nuclear transfer Abandoned US20070033664A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/543,786 US20070033664A1 (en) 1995-08-31 2006-10-06 Quiescent cell populations for nuclear transfer

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GBGB9517780.4A GB9517780D0 (en) 1995-08-31 1995-08-31 Biological manipulation
GBGB9517780.4 1995-08-31
PCT/GB1996/002099 WO1997007669A1 (en) 1995-08-31 1996-08-30 Quiescent cell populations for nuclear transfer
US08/802,282 US6147276A (en) 1995-08-31 1997-02-19 Quiescent cell populations for nuclear transfer in the production of non-human mammals and non-human mammalian embryos
US22523399A 1999-01-04 1999-01-04
US10/915,338 US7232938B2 (en) 1995-08-31 2004-08-11 Cloning ungulates from a quiescent donor cell
US11/543,786 US20070033664A1 (en) 1995-08-31 2006-10-06 Quiescent cell populations for nuclear transfer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/915,338 Continuation US7232938B2 (en) 1995-08-31 2004-08-11 Cloning ungulates from a quiescent donor cell

Publications (1)

Publication Number Publication Date
US20070033664A1 true US20070033664A1 (en) 2007-02-08

Family

ID=10779998

Family Applications (5)

Application Number Title Priority Date Filing Date
US08/802,282 Expired - Lifetime US6147276A (en) 1995-08-31 1997-02-19 Quiescent cell populations for nuclear transfer in the production of non-human mammals and non-human mammalian embryos
US10/915,338 Expired - Fee Related US7232938B2 (en) 1995-08-31 2004-08-11 Cloning ungulates from a quiescent donor cell
US11/265,113 Expired - Fee Related US7514258B2 (en) 1995-08-31 2005-11-03 Mammalian cultured inner cell mass cell culture using a quiescent cell as nuclear
US11/543,786 Abandoned US20070033664A1 (en) 1995-08-31 2006-10-06 Quiescent cell populations for nuclear transfer
US11/544,039 Expired - Fee Related US7355094B2 (en) 1995-08-31 2006-10-06 Methods of ungulate nuclear transfer

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/802,282 Expired - Lifetime US6147276A (en) 1995-08-31 1997-02-19 Quiescent cell populations for nuclear transfer in the production of non-human mammals and non-human mammalian embryos
US10/915,338 Expired - Fee Related US7232938B2 (en) 1995-08-31 2004-08-11 Cloning ungulates from a quiescent donor cell
US11/265,113 Expired - Fee Related US7514258B2 (en) 1995-08-31 2005-11-03 Mammalian cultured inner cell mass cell culture using a quiescent cell as nuclear

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/544,039 Expired - Fee Related US7355094B2 (en) 1995-08-31 2006-10-06 Methods of ungulate nuclear transfer

Country Status (24)

Country Link
US (5) US6147276A (en)
EP (3) EP0930009A1 (en)
JP (2) JP4081613B2 (en)
KR (4) KR20060053008A (en)
CN (3) CN100422317C (en)
AT (1) ATE199115T1 (en)
AU (1) AU716956C (en)
BR (1) BR9610034B1 (en)
CA (1) CA2229568C (en)
CZ (1) CZ60898A3 (en)
DE (1) DE69611793T2 (en)
DK (1) DK0849990T3 (en)
ES (1) ES2155197T3 (en)
GB (2) GB9517780D0 (en)
HK (2) HK1004938A1 (en)
HU (1) HUP9900234A3 (en)
IL (1) IL123298A0 (en)
NO (1) NO980845L (en)
NZ (3) NZ337311A (en)
PL (1) PL325331A1 (en)
SG (1) SG75155A1 (en)
SI (1) SI0849990T1 (en)
WO (1) WO1997007669A1 (en)
ZA (1) ZA967390B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050172347A1 (en) * 1995-08-31 2005-08-04 Campbell Keith H.S. Unactivated oocytes as cytoplast recipients for nuclear transfer
US20060156424A1 (en) * 1995-08-31 2006-07-13 Roslin Institute (Edinburgh); Minister of Agriculture, Fisheries & Food Quiescent cell populations for nuclear transfer

Families Citing this family (221)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5480772A (en) 1993-02-03 1996-01-02 Brandeis University In vitro activation of a nucleus
US7304204B2 (en) 1995-08-31 2007-12-04 Roslin Institute Ungulates produced by nuclear transfer of G1 cells
US6077697A (en) 1996-04-10 2000-06-20 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US6025155A (en) * 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
US7696404B2 (en) 1996-08-19 2010-04-13 Advanced Cell Technology, Inc. Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
WO1998016630A1 (en) 1996-10-11 1998-04-23 The Texas A & M University System Methods for the generation of primordial germ cells and transgenic animal species
US6215041B1 (en) * 1997-01-10 2001-04-10 University Of Mmassachusetts Cloning using donor nuclei from a non-quiesecent somatic cells
US6235969B1 (en) * 1997-01-10 2001-05-22 University Of Massachusetts Cloning pigs using donor nuclei from non-quiescent differentiated cells
US20090092588A1 (en) * 1997-01-10 2009-04-09 University Of Massachusetts As Represented By Its Amherst Campus Cloned ungulate embryos and animals, use of cells, tissues and organs thereof for transplantation therapies including parkinson's disease
US5945577A (en) * 1997-01-10 1999-08-31 University Of Massachusetts As Represented By Its Amherst Campus Cloning using donor nuclei from proliferating somatic cells
GB9703550D0 (en) * 1997-02-20 1997-04-09 Ppl Therapeutics Scotland Ltd Cell mediated transgenesis
US6011197A (en) 1997-03-06 2000-01-04 Infigen, Inc. Method of cloning bovines using reprogrammed non-embryonic bovine cells
CA2289277C (en) 1997-04-24 2013-02-12 University Of Washington Targeted gene modification by parvoviral vectors
WO1998057984A2 (en) 1997-06-19 1998-12-23 The General Hospital Corporation Torsin, torsin genes, and methods of use
US6921814B2 (en) 1997-06-19 2005-07-26 The General Hospital Corporation Torsin, torsin-related genes and methods of detecting neuronal disease
BR9811657A (en) * 1997-07-03 2000-09-05 Univ Massachusetts Public Inst Cloning using donor nuclei from differentiated serum-free cells
WO1999005266A2 (en) * 1997-07-26 1999-02-04 Wisconsin Alumni Research Foundation Trans-species nuclear transfer
AU8817798A (en) * 1997-08-22 1999-03-16 Biotechnology And Biological Sciences Research Council, The Use of (mariner) transposan in the production of transgenic animals
AU1581899A (en) * 1997-10-28 1999-05-17 Biotransplant Incorporated Nuclear transfer for production of transgenic animal embryo
RU2216592C2 (en) * 1998-01-16 2003-11-20 Агробиоген Гмбх Method for obtaining animal embryos and method for raising animals out of embryos
US6331659B1 (en) * 1998-01-21 2001-12-18 University Of Hawaii Cumulus cells as nuclear donors
JP2002511234A (en) * 1998-03-16 2002-04-16 リレグ ピーティーワイ リミテッド Swine nuclear transfer
AUPP294898A0 (en) * 1998-04-15 1998-05-07 Monash University Method of nuclear transfer
GB9808325D0 (en) * 1998-04-20 1998-06-17 Ltr Ciz Di Associazione Italia Source of nuclei for nuclear transfer
GB9809178D0 (en) * 1998-04-29 1998-07-01 Univ Edinburgh Nuclear reprogramming of somatic cells
GB2365445A (en) * 1998-10-12 2002-02-20 Geron Bio Med Ltd Porcine occcytes with improved developmental competence
EP1375654A3 (en) * 1998-11-02 2008-01-16 GTC Biotherapeutics, Inc. Transgenic and cloned mammals
US6580017B1 (en) 1998-11-02 2003-06-17 Genzyme Transgenics Corporation Methods of reconstructed goat embryo transfer
AU2003204830B2 (en) * 1998-11-02 2007-08-30 Gtc Biotherapeutics, Inc. Transgenic and cloned mammals
EP1127113A2 (en) * 1998-11-02 2001-08-29 Genzyme Transgenics Corporation Transgenic and cloned mammals
US6781030B1 (en) 1998-11-02 2004-08-24 Trustee Of Tufts College, Ballou Hall Methods for cloning mammals using telophase oocytes
WO2000030436A1 (en) 1998-11-19 2000-06-02 Ppl Therapeutics (Scotland) Ltd. Stabilisation of milk from transgenic animals
US6258998B1 (en) 1998-11-24 2001-07-10 Infigen, Inc. Method of cloning porcine animals
US6700037B2 (en) 1998-11-24 2004-03-02 Infigen, Inc. Method of cloning porcine animals
WO2000042174A1 (en) * 1999-01-13 2000-07-20 Ppl Therapeutics (Scotland) Limited Double nuclear transfer method and results thereof
GB9903805D0 (en) * 1999-02-20 1999-04-14 Univ Sheffield Pluripotential cells-1
GB9903804D0 (en) * 1999-02-20 1999-04-14 Univ Sheffield Pluripotential cells-2
WO2000050443A2 (en) 1999-02-26 2000-08-31 Millennium Pharmaceutcals, Inc. Secreted proteins and uses thereof
CA2361943C (en) 1999-03-04 2011-05-17 Ppl Therapeutics (Scotland) Limited Genetic modification of somatic cells and uses thereof
US20020012660A1 (en) * 1999-03-04 2002-01-31 Alan Colman Method of preparing a somatic cells for nuclear transfer
WO2000065035A1 (en) * 1999-04-28 2000-11-02 Universite De Montreal Telophase enucleated oocytes for nuclear transfer
DE69934914T2 (en) * 1999-06-04 2007-10-18 Istituto Zootecnico E Caseario Per La Sardegna METHOD FOR THE RECONSTRUCTION OF NON-HUMAN ANIMAL EMBRYOS BY CORE TRANSFER AND PRODUCTION OF THE ANIMAL OUT OF THESE.
WO2001000855A1 (en) 1999-06-23 2001-01-04 Ppl Therapeutics (Scotland) Ltd. Fusion proteins incorporating lysozyme
EP1117763A4 (en) * 1999-06-30 2004-12-01 Hwang Woo Suk Method for producing cloned tigers by employing inter-species nuclear transplantation technique
WO2001000795A1 (en) * 1999-06-30 2001-01-04 Hwang, Woo-Suk A method for producing cloned cows
JP2003503071A (en) 1999-06-30 2003-01-28 アドバンスド セル テクノロジー、インコーポレイテッド Cytoplasmic transfer to dedifferentiated recipient cells
WO2001000793A1 (en) * 1999-06-30 2001-01-04 Hwang, Woo-Suk Method for producing human cloned embryos by employing inter-species nuclear transplantation technique
US7291714B1 (en) 1999-06-30 2007-11-06 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
DK1210428T3 (en) 1999-08-23 2015-06-15 Dana Farber Cancer Inst Inc PD-1, a receptor for B7-4 AND USE THEREOF
WO2002064799A2 (en) 1999-09-28 2002-08-22 Transkaryotic Therapies, Inc. Optimized messenger rna
EP2275559A3 (en) 1999-09-28 2011-03-23 Shire Human Genetic Therapies, Inc. Optimized messenger RNA
BR0014448A (en) * 1999-09-30 2002-09-17 Genzyme Transgenics Corp Methods of production of cloned and transgenic mammals
AU1777301A (en) * 1999-11-19 2001-05-30 Hematech, Llc Production of ungulates, preferably bovines that produce human immunoglobulins
US7414170B2 (en) * 1999-11-19 2008-08-19 Kirin Beer Kabushiki Kaisha Transgenic bovines capable of human antibody production
US7074983B2 (en) 1999-11-19 2006-07-11 Kirin Beer Kabushiki Kaisha Transgenic bovine comprising human immunoglobulin loci and producing human immunoglobulin
US7820878B2 (en) * 1999-11-19 2010-10-26 Kyowa Hakko Kirin Co., Ltd. Production of ungulates, preferably bovines that produce human immunoglobulins
KR100417566B1 (en) * 1999-11-19 2004-02-05 한국생명공학연구원 A mass production method of embryos by nuclear transfer using somatic cells
US6635802B1 (en) * 2000-01-10 2003-10-21 The Texas A&M University System Nuclear transfer using cells cultured in serum starvation media containing apoptosis inhibitors
GB0001401D0 (en) * 2000-01-22 2000-03-08 Univ Edinburgh Methods for amplifying genetic material and use thereof
EP1356031B1 (en) 2000-03-15 2011-07-27 The University Of Georgia Research Foundation, Inc. Effective nuclear reprogramming in mammals
US20040033600A1 (en) 2001-03-21 2004-02-19 Palli Subba Reddy Ecdysone receptor-based inducible gene expression system
GB2360522A (en) 2000-03-24 2001-09-26 Geron Corp A strategy for maintaining pregnancy
WO2001088096A2 (en) * 2000-05-15 2001-11-22 Geron Corporation Ovine tissue for xenotransplantation
JP2004501624A (en) 2000-06-28 2004-01-22 ジェネティックス・インスチチュート・リミテッド・ライアビリティ・カンパニー PD-L2 molecule: novel PD-1 ligand and use thereof
WO2002009510A2 (en) * 2000-08-01 2002-02-07 Xy Genetics, Llc Methods of selecting and cloning animals
CA2422155A1 (en) 2000-08-03 2002-02-14 Wim Van Schooten Production of humanized antibodies in transgenic animals
US6677500B2 (en) 2000-08-15 2004-01-13 Board Of Trustees Of The University Of Illinois Ungulates expressing exogenous IGF-I in their milk
WO2002019811A2 (en) * 2000-09-06 2002-03-14 Nexia Biotechnologies, Inc. Generation of transgenic animals using nuclear transfer and oocytes at the germinal vesicle stage
US8105825B2 (en) * 2000-10-03 2012-01-31 Intrexon Corporation Multiple inducible gene regulation system
US20040029825A1 (en) * 2001-10-03 2004-02-12 Davies Christopher J Methods of minimizing immunological rejection of a nuclear transfer fetus
FR2814642B1 (en) 2000-10-03 2005-07-01 Ass Pour Le Dev De La Rech En TRANSGENIC MOUSE FOR THE TARGETED RECOMBINATION MEDIATED BY THE MODIFIED CRE-ER
EP1356108A2 (en) 2000-11-28 2003-10-29 Wyeth Expression analysis of fkbp nucleic acids and polypeptides useful in the diagnosis and treatment of prostate cancer
EP1343912A2 (en) 2000-11-28 2003-09-17 Wyeth Expression analysis of kiaa nucleic acids and polypeptides useful in the diagnosis and treatment of prostate cancer
US20020142397A1 (en) 2000-12-22 2002-10-03 Philippe Collas Methods for altering cell fate
US7491534B2 (en) 2000-12-22 2009-02-17 Kirin Holdings Kabushiki Kaisha Methods for altering cell fate to generate T-cells specific for an antigen of interest
EP2138583A1 (en) 2000-12-22 2009-12-30 Institut National De La Recherche Agronomique Position-independent and tissue specific expression of a transgene in milk of transgenic animals
JP4420604B2 (en) * 2000-12-22 2010-02-24 協和発酵キリン株式会社 Methods for cloning mammals using reprogrammed donor chromatin or donor cells
EP1860199B1 (en) 2001-01-12 2014-03-05 Exelixis, Inc. Modulating insulin receptor signaling
WO2002074078A2 (en) 2001-01-30 2002-09-26 Rebholtz, Amy, K. Method for embryo after impregnation of the recipient female
WO2002062131A2 (en) * 2001-02-02 2002-08-15 Infigen, Inc. Method of cloning transgenic mammalian animals using pseudonuclei
ES2392508T3 (en) * 2001-02-20 2012-12-11 Intrexon Corporation Chimeric X retinoid receptors and their use in an inducible gene expression system based on novel ecdysone receptors
EP1434988A4 (en) 2001-02-23 2005-12-14 Elan Pharm Inc Transgenic knockouts of bace-1
JP2002262716A (en) * 2001-03-07 2002-09-17 National Agricultural Research Organization Method for creating livestock individual using established cell line
WO2002072762A2 (en) * 2001-03-08 2002-09-19 Advanced Cell Technology, Inc. Use of rna interference for the creation of lineage specific es and other undifferentiated cells and production of differentiated cells in vitro by co-culture
AUPR363701A0 (en) 2001-03-09 2001-04-05 Garelag Pty Ltd Method for the production of nuclear transfer embryos
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US7126039B2 (en) * 2001-03-21 2006-10-24 Geron Corporation Animal tissue with carbohydrate antigens compatible for human transplantation
US7265262B2 (en) * 2001-03-21 2007-09-04 Roslin Institute (Edinburgh) Telomerizing nuclear donor cells and improving the efficiency on nuclear transfer
US20030027330A1 (en) * 2001-04-02 2003-02-06 Robert Lanza Method for facilitating the production of differentiated cell types and tissues from embryonic and adult pluripotent and multipotent cells
WO2002079499A1 (en) 2001-04-02 2002-10-10 Wyeth Pd-1, a receptor for b7-4, and uses therefor
EP2258842A1 (en) 2001-04-16 2010-12-08 Wyeth Holdings Corporation Streptococcus pneumoniae open reading frames encoding polypeptide antigens and uses thereof
US20030153044A1 (en) * 2001-05-14 2003-08-14 Monika Liljedahl Tissues or organs for use in xenotransplantation
WO2002092791A1 (en) * 2001-05-14 2002-11-21 Stell Tissues or organs for use in xenotransplantation
WO2003005810A2 (en) * 2001-07-09 2003-01-23 Alexion Pharmaceuticals Inc. Cloned non-human mammals from contact inhibited donor cells
EP1900815B1 (en) 2001-07-12 2016-09-07 University of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
ES2566561T3 (en) 2001-07-12 2016-04-13 University Of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
US20030051264A1 (en) * 2001-07-31 2003-03-13 Monika Liljedahl Genetically modified cows having reduced susceptibility to mad cow disease
US20030211603A1 (en) * 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
WO2003042362A2 (en) 2001-11-09 2003-05-22 Dana-Farber Cancer Institute, Inc. PGC-1β, A NOVEL PGC-1 HOMOLOGUE AND USES THEREFOR
US20050090004A1 (en) * 2003-01-16 2005-04-28 Sayre Chauncey B. Stem cell maturation for all tissue lines
US20030134422A1 (en) * 2002-01-16 2003-07-17 Sayre Chauncey Bigelow Stem cell maturation for all tissue lines
US20050170506A1 (en) * 2002-01-16 2005-08-04 Primegen Biotech Llc Therapeutic reprogramming, hybrid stem cells and maturation
BRPI0307383B1 (en) * 2002-01-23 2019-12-31 The Univ Of Utah Research Foundation directed genetic recombination method in host plant cell
KR100502695B1 (en) * 2002-02-28 2005-07-25 한국생명공학연구원 A method for mass production of cloned embryos for improving transgenesis in animals
ATE531796T1 (en) * 2002-03-21 2011-11-15 Sangamo Biosciences Inc METHODS AND COMPOSITIONS FOR USING ZINC FINGER ENDONUCLEASES TO IMPROVE HOMOLOGOUS RECOMBINATION
GB0207299D0 (en) * 2002-03-28 2002-05-08 Franks Christopher R Stem cell therapy
CA2480802A1 (en) * 2002-04-01 2003-10-16 Gtc Biotherapeutics, Inc. A method for selecting cell lines to be used for nuclear transfer in mammalian species
US20030207448A1 (en) * 2002-05-06 2003-11-06 Revera Gregory Henry Methodologies for the creation of pluripotent or multipotent human stem cells without creating or destroying a human embryo
GB0211904D0 (en) * 2002-05-23 2002-07-03 Medical Res Council Nuclear transfer
AU2003247514A1 (en) * 2002-06-11 2003-12-22 Roy Ogle Meningeal-derived stem cells
US7612250B2 (en) * 2002-07-29 2009-11-03 Trustees Of Tufts College Nuclear transfer embryo formation method
KR20050058407A (en) 2002-08-20 2005-06-16 밀레니엄 파머슈티컬스 인코퍼레이티드 Compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
US9447434B2 (en) * 2002-09-05 2016-09-20 California Institute Of Technology Use of chimeric nucleases to stimulate gene targeting
EP1575518A4 (en) 2002-10-10 2007-08-22 Wyeth Corp Compositions, organisms and methodologies employing a novel human kinase
US20040077077A1 (en) * 2002-10-18 2004-04-22 Xiangzhong Yang Novel methods for the production of cloned mammals, mammals cloned according to the methods, and methods of use of same
US7208306B2 (en) 2002-10-24 2007-04-24 Wyeth Compositions employing a novel human protein phosphatase
US7429690B2 (en) 2002-11-08 2008-09-30 Kirin Holdings Kabushiki Kaisha Transgenic bovines having reduced prion protein production
BR0316111A (en) 2002-11-21 2005-09-13 Wyeth Corp Methods to diagnose rcc and other solid tumors
US7297525B2 (en) 2002-11-27 2007-11-20 Wyeth Composition employing a novel human kinase
JP2006508676A (en) * 2002-12-10 2006-03-16 ジーティーシー バイオセラピューティックス インコーポレイテッド Methods and systems utilizing somatic cell nuclear transfer embryos as cell donors for additional nuclear transfer
EP2151507B1 (en) 2002-12-20 2013-02-13 Celera Corporation Genetic polymorphisms associated with myocardial infarction, methods of detection and uses thereof
EP1998177A3 (en) 2003-01-06 2009-02-18 Wyeth Compositions and methods for diagnosing and treating colon cancers
FR2855183B1 (en) 2003-05-23 2005-08-12 Agronomique Inst Nat Rech PROCESS FOR CLONING THE RAT BY NUCLEAR TRANSFER
CA2528012C (en) 2003-06-02 2015-11-24 University Of Massachusetts Methods and compositions for controlling efficacy of rna silencing
ES2864206T3 (en) 2003-06-02 2021-10-13 Univ Massachusetts Methods and compositions to improve the efficacy and specificity of RNAi
US20120196370A1 (en) 2010-12-03 2012-08-02 Fyodor Urnov Methods and compositions for targeted genomic deletion
US11311574B2 (en) 2003-08-08 2022-04-26 Sangamo Therapeutics, Inc. Methods and compositions for targeted cleavage and recombination
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
US8409861B2 (en) * 2003-08-08 2013-04-02 Sangamo Biosciences, Inc. Targeted deletion of cellular DNA sequences
AU2004265662B2 (en) 2003-08-14 2010-12-16 Exelixis, Inc. UPs as modifiers of the beta catenin pathway and methods of use
CA2761987A1 (en) 2003-10-07 2005-04-21 Millennium Pharmaceuticals, Inc. Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer
EP1681988B1 (en) 2003-11-12 2015-04-08 The Children's Hospital Medical Center Method for diagnosis and treatment of pulmonary disorders
CA2547072C (en) 2003-11-26 2015-06-23 Applera Corporation Single nucleotide polymorphisms associated with cardiovascular disorders and statin response, methods of detection and uses thereof
US7972854B2 (en) 2004-02-05 2011-07-05 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
WO2005104835A2 (en) 2004-04-22 2005-11-10 Kirin Beer Kabushiki Kaisha Transgenic animals and uses thereof
WO2005111241A2 (en) 2004-05-07 2005-11-24 Applera Corporation Genetic polymorphisms associated with liver fibrosis methods of detection and uses thereof
JP2006081542A (en) * 2004-08-18 2006-03-30 Institute Of Physical & Chemical Research Method for creating clone mammal
WO2006047312A2 (en) 2004-10-22 2006-05-04 Dana-Farber Cancer Institute, Inc. COMPOSITIONS AND METHODS FOR MODULATING PGC-1β TO TREAT LIPID-RELATED DISEASES AND DISORDERS
GB0426146D0 (en) 2004-11-29 2004-12-29 Bioxell Spa Therapeutic peptides and method
CN1274829C (en) 2004-12-10 2006-09-13 四川大学华西医院 Anti EB virus resulted tumour polypeptide, and its use and preparing method
EP1856296A2 (en) 2005-03-11 2007-11-21 Applera Corporation Genetic polymorphisms associated with coronary heart disease, methods of detection and uses thereof
US20060286666A1 (en) * 2005-06-17 2006-12-21 Kang Ji Y Methods and apparatuses for culturing stem cell using biomaterial shell
WO2007052096A2 (en) * 2005-06-29 2007-05-10 Cameron Malcolm Lang Clokie Production of bone morphogenic proteins (bmps) in transgenic mammals
KR100733012B1 (en) 2005-07-26 2007-06-28 재단법인서울대학교산학협력재단 Cloned Caninds And Method For Producing Thereof
AU2006292827B2 (en) 2005-08-09 2013-02-14 Revivicor, Inc. Transgenic ungulates expressing CTLA4-IG and uses thereof
WO2007030820A2 (en) 2005-09-09 2007-03-15 The Johns Hopkins University Manipulation of regulatory t cell and dc function by targeting neuritin gene using antibodies, agonists and antagonists
EP1926459B1 (en) 2005-09-19 2015-01-07 Histogenics Corporation Cell-support matrix having narrowly defined uniformly vertically and non-randomly organized porosity and pore density and a method for preparation thereof
US7799530B2 (en) 2005-09-23 2010-09-21 Celera Corporation Genetic polymorphisms associated with cardiovascular disorders and drug response, methods of detection and uses thereof
US7695911B2 (en) 2005-10-26 2010-04-13 Celera Corporation Genetic polymorphisms associated with Alzheimer's Disease, methods of detection and uses thereof
WO2007124019A2 (en) 2006-04-21 2007-11-01 Gtc Biotherapeutics, Inc. Methods and products related to the transfer of molecules from blood to the mammary gland
CN102719444B (en) 2006-09-01 2016-12-14 人类多细胞株治疗学公司 The expression that people or Humanized immunoglobulin strengthen in non-human transgenic animal
EP2639317B1 (en) 2006-09-11 2015-03-04 Celera Corporation Genetic polymorphisms associated with psoriasis, methods of detection and uses thereof
WO2008051511A2 (en) 2006-10-20 2008-05-02 Applera Corporation Genetic polymorphisms associated with venous thrombosis, methods of detection and uses thereof
KR101398220B1 (en) * 2006-10-30 2014-05-22 건국대학교 산학협력단 Methods for producing transgenic embryos
US20100205678A1 (en) * 2007-01-05 2010-08-12 Overstrom Eric W Oocyte spindle-associated factors improve somatic cell cloning
KR100829426B1 (en) 2007-01-17 2008-05-15 재단법인서울대학교산학협력재단 Method for producing cloned dog
US20080222745A1 (en) * 2007-03-07 2008-09-11 Utah State University Colcemid-Treatment of Oocytes to enhance Nuclear Transfer Cloning
KR100868245B1 (en) 2007-05-29 2008-11-11 강원대학교산학협력단 Effective producing method of cloned animal by demecolcine treatment
EP2214708A4 (en) 2007-10-26 2011-01-12 Centocor Ortho Biotech Inc Vectors, host cells, and methods of production and uses
US8039212B2 (en) 2007-11-05 2011-10-18 Celera Corporation Genetic polymorphisms associated with liver fibrosis, methods of detection and uses thereof
JP2011508600A (en) 2008-01-04 2011-03-17 ソウル ナショナル ユニバーシティ インダストリー ファウンデーション Method for producing transformed replicated dogs
US20090221620A1 (en) 2008-02-20 2009-09-03 Celera Corporation Gentic polymorphisms associated with stroke, methods of detection and uses thereof
WO2009114185A2 (en) 2008-03-12 2009-09-17 The Rockefeller University Methods and compositions for translational profiling and molecular phenotyping
US8288094B2 (en) 2008-04-09 2012-10-16 Institut Pasteur APOBEC3 mediated DNA editing
US8557773B2 (en) 2008-05-02 2013-10-15 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Treatment of bleeding with low half-life fibrinogen
DK2295538T3 (en) 2008-07-07 2015-12-21 Takara Bio Inc Process for producing a pluripotent stem cell
US8216786B2 (en) 2008-07-09 2012-07-10 Celera Corporation Genetic polymorphisms associated with cardiovascular diseases, methods of detection and uses thereof
US9181315B2 (en) 2009-01-08 2015-11-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for induced brown fat differentiation
US8722964B2 (en) 2009-04-23 2014-05-13 Transposagen Biopharmaceuticals, Inc. Genetically engineered or transgenic rats exhibiting a cancer phenotype due to a disruption of germline tumor suppressor genes
EP2267153A1 (en) 2009-05-26 2010-12-29 Université Claude Bernard Lyon 1 Identification of netrin-1 receptor unc5c gene mutation in solid cancers
WO2011002988A1 (en) 2009-07-01 2011-01-06 Transposagen Biopharmaceuticals, Inc. Genetically modified rat models for severe combined immunodeficiency (scid)
WO2011011678A2 (en) 2009-07-24 2011-01-27 Transposagen Biopharmaceuticals, Inc. Genetically modified rat models for cytokine-cytokine signaling pathways
WO2011014721A2 (en) 2009-07-30 2011-02-03 Transposagen Biopharmaceuticals, Inc. Genetically modified rat models for pharmacokinetics
WO2011017518A2 (en) 2009-08-05 2011-02-10 Transposagen Biopharmaceuticals, Inc. Genetically modified rat models for drug metabolism
WO2011022634A2 (en) 2009-08-20 2011-02-24 Transposagen Biopharmaceuticals, Inc. Genetically modified rat models for pain
US9420770B2 (en) 2009-12-01 2016-08-23 Indiana University Research & Technology Corporation Methods of modulating thrombocytopenia and modified transgenic pigs
US10429384B2 (en) 2010-01-22 2019-10-01 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of metabolic disorders
TWI518325B (en) 2010-02-04 2016-01-21 自治醫科大學 Identification, assessment, and therapy of cancers with innate or acquired resistance to alk inhibitors
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
AU2011230537C1 (en) 2010-03-26 2018-08-02 Trustees Of Dartmouth College Vista regulatory T cell mediator protein, vista binding agents and use thereof
US8383793B2 (en) 2010-04-15 2013-02-26 St. Jude Children's Research Hospital Methods and compositions for the diagnosis and treatment of cancer resistant to anaplastic lymphoma kinase (ALK) kinase inhibitors
US20110269735A1 (en) 2010-04-19 2011-11-03 Celera Corporation Genetic polymorphisms associated with statin response and cardiovascular diseases, methods of detection and uses thereof
ES2738476T3 (en) 2010-08-11 2020-01-23 Inst Nat Sante Rech Med Cocultures of cluster cells and embryos during in vitro fertilization procedures
US20120108651A1 (en) 2010-11-02 2012-05-03 Leiden University Medical Center (LUMC) Acting on Behalf of Academic Hospital Leiden (AZL) Genetic polymorphisms associated with venous thrombosis and statin response, methods of detection and uses thereof
WO2012071393A2 (en) 2010-11-23 2012-05-31 The New York Stem Cell Foundation Method for producing pluripotent stem cells
CN104774871A (en) 2011-02-14 2015-07-15 雷维维科公司 Genetically modified pigs for xenotransplantation of vascularized xenografts and derivatives thereof
WO2012129198A1 (en) 2011-03-23 2012-09-27 Transposagen Biopharmaceuticals, Inc. Genetically modified rat models for obesity and diabetes
KR102574897B1 (en) 2011-05-16 2023-09-06 더 큐레이터스 오브 더 유니버시티 오브 미주리 Porcine reproductive and respiratory syndrome virus resistant animals
US10093705B2 (en) 2011-09-13 2018-10-09 Dana-Farber Cancer Institute, Inc. Compositions and methods for brown fat induction and activity using FNDC5
CN102391989A (en) * 2011-10-12 2012-03-28 扬州大学 Exact analysis inspection method for transgenic animal recombinant product
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
SI2814842T1 (en) 2012-02-15 2018-10-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
PL2814844T3 (en) 2012-02-15 2017-12-29 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (trem-1)
TWI677507B (en) 2012-06-22 2019-11-21 達特茅斯學院基金會 Novel vista-ig constructs and the use of vista-ig for treatment of autoimmune, allergic and inflammatory disorders
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
EP3552628A1 (en) 2012-09-07 2019-10-16 The Trustees Of Dartmouth College Vista modulators for diagnosis and treatment of cancer
US20140115728A1 (en) 2012-10-24 2014-04-24 A. Joseph Tector Double knockout (gt/cmah-ko) pigs, organs and tissues
US10034921B2 (en) 2013-02-13 2018-07-31 Laboratoire Français Du Fractionnement Et Des Biotechnologies Proteins with modified glycosylation and methods of production thereof
JP2016508515A (en) 2013-02-13 2016-03-22 ラボラトワール フランセ デュ フラクショヌマン エ デ ビオテクノロジーLaboratoire Francais du Fractionnement et des Biotechnologies Highly galactosylated anti-TNF-α antibody and use thereof
PL2993234T3 (en) 2013-04-30 2019-04-30 Univ Konkuk Ind Coop Corp Cmp-acetylneuraminic acid hydroxylase targeting vector, transgenic animal for xenotransplantation introduced with the vector, and method of manufacturing the same
MX369173B (en) 2013-12-24 2019-10-30 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments.
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
CN106536559B (en) 2014-07-17 2021-04-27 诺和诺德股份有限公司 Site-directed mutagenesis of TREM-1 antibodies to reduce viscosity
US10077420B2 (en) 2014-12-02 2018-09-18 Histogenics Corporation Cell and tissue culture container
CA2969730A1 (en) 2014-12-05 2016-06-09 Immunext, Inc. Identification of vsig8 as the putative vista receptor and its use thereof to produce vista/vsig8 modulators
HUE052606T2 (en) 2014-12-09 2021-05-28 Regeneron Pharma Non-human animals having a humanized cluster of differentiation 274 gene
CN104823921A (en) * 2015-05-21 2015-08-12 蒙汉勤 Feeding technology of mini fragrant pigs for human body burnt skin transplantation and medical experiments
MX2017016647A (en) 2015-06-24 2019-04-25 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments.
US10091975B2 (en) 2015-08-06 2018-10-09 The Curators Of The University Of Missouri Pathogen-resistant animals having modified CD163 genes
US20210106661A1 (en) 2015-10-29 2021-04-15 Dana-Farber Cancer Institute, Inc. Methods for identification, assessment, prevention, and treatment of metabolic disorders using pm20d1 and n-lipidated amino acids
CN116920085A (en) 2016-02-12 2023-10-24 詹森药业有限公司 anti-VISTA (B7H 5) antibodies
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
SG11202009625WA (en) 2018-04-02 2020-10-29 Bristol Myers Squibb Co Anti-trem-1 antibodies and uses thereof
JP2021520781A (en) 2018-04-06 2021-08-26 チルドレンズ メディカル センター コーポレーションChildren’S Medical Center Corporation Compositions and Methods for Somatic Cell Reprogramming and Imprinting Modulation
US11160260B2 (en) 2018-04-17 2021-11-02 The Curators Of The University Of Missouri Methods for protecting porcine fetuses from infection with porcine reproductive and respiratory syndrome virus (PRRSV)
WO2020041360A1 (en) 2018-08-21 2020-02-27 Quidel Corporation Dbpa antibodies and uses thereof
EP4077376A2 (en) 2019-12-19 2022-10-26 Quidel Corporation Monoclonal antibody fusions
AU2021385081A1 (en) 2020-11-20 2023-06-29 Revivicor, Inc. Multi-transgenic pigs with growth hormone receptor knockout for xenotransplantation
WO2023044100A1 (en) 2021-09-20 2023-03-23 Revivicor, Inc. Multitran scenic pigs comprising ten genetic modifications for xenotransplantation

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057420A (en) * 1987-06-05 1991-10-15 Granada Biosciences, Inc. Bovine nuclear transplantation
US5496720A (en) * 1993-02-10 1996-03-05 Susko-Parrish; Joan L. Parthenogenic oocyte activation
US5523226A (en) * 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US5945577A (en) * 1997-01-10 1999-08-31 University Of Massachusetts As Represented By Its Amherst Campus Cloning using donor nuclei from proliferating somatic cells
US6147276A (en) * 1995-08-31 2000-11-14 Roslin Institute (Edinburgh) Quiescent cell populations for nuclear transfer in the production of non-human mammals and non-human mammalian embryos
US6215041B1 (en) * 1997-01-10 2001-04-10 University Of Mmassachusetts Cloning using donor nuclei from a non-quiesecent somatic cells
US6235969B1 (en) * 1997-01-10 2001-05-22 University Of Massachusetts Cloning pigs using donor nuclei from non-quiescent differentiated cells
US6252133B1 (en) * 1995-08-31 2001-06-26 Roslin Institute (Edinburgh) Unactivated oocytes as cytoplast recipients of quiescent and non-quiescent cell nuclei, while maintaining correct ploidy
US6548741B2 (en) * 1998-10-12 2003-04-15 Geron Corporation Developmental competence for assisted reproduction and nuclear transfer in pigs
US6603059B1 (en) * 1997-03-06 2003-08-05 Infigen, Inc. Method of cloning animals

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR890003947B1 (en) * 1985-12-11 1989-10-13 가부시기가이샤 시마즈세이사구쇼 Apparatus for cell fusion
GB8714426D0 (en) * 1987-06-19 1987-07-22 Agricultural & Food Res Culture technique
CA2059199A1 (en) * 1992-01-10 1993-07-11 Steen Willadsen Method for producing mature reconstituted mammalian eggs by nuclear transplantation
GB2265909B (en) * 1992-04-01 1996-03-20 Inst Of Animal Physiology And Fusion of donor nucleus with recipient cytoplast in the absence of calcium ion
CA2145190A1 (en) * 1992-09-22 1994-03-31 The Government Of The United States Of America As Represented By The Sec Retary, Department Of Health And Human Services Use of taxol for treating lymphomas and breast cancer
CA2092258A1 (en) * 1992-12-30 1994-07-01 Steven L. Stice Synchronized donor cells for bovine nuclear transfer
DE69433381T3 (en) * 1993-07-29 2009-04-16 The Government of the United States of America, as represented by the Secretary National Institute of Health, Office of Technology Transfer METHOD FOR TREATING ATHEROSCLEROSIS OR RESTENOSIS WITH THE HELP OF A MICROTUBULUS STABILIZER
WO1995016770A1 (en) * 1993-12-17 1995-06-22 Abs Global, Inc. Ungulate preblastocyst derived embryonic stem cells and use thereof to produce cloned transgenic and chimeric ungulates
EP1149898A2 (en) * 1993-12-23 2001-10-31 Infigen, Inc. Embryonic stem cells as nuclear donors and nuclear transfer techniques to produce chimeric and transgenic animals
GB9417831D0 (en) * 1994-09-05 1994-10-26 Biotech & Biolog Scien Res Biological manipulation
US7304204B2 (en) 1995-08-31 2007-12-04 Roslin Institute Ungulates produced by nuclear transfer of G1 cells
WO1999005266A2 (en) * 1997-07-26 1999-02-04 Wisconsin Alumni Research Foundation Trans-species nuclear transfer
US6225243B1 (en) * 1998-08-03 2001-05-01 Bba Nonwovens Simpsonville, Inc. Elastic nonwoven fabric prepared from bi-component filaments
US20050156424A1 (en) * 2000-02-29 2005-07-21 Thyssenkrupp Presta Ag Steering column and manufacturing method thereof

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057420A (en) * 1987-06-05 1991-10-15 Granada Biosciences, Inc. Bovine nuclear transplantation
US5496720A (en) * 1993-02-10 1996-03-05 Susko-Parrish; Joan L. Parthenogenic oocyte activation
US5523226A (en) * 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US6147276A (en) * 1995-08-31 2000-11-14 Roslin Institute (Edinburgh) Quiescent cell populations for nuclear transfer in the production of non-human mammals and non-human mammalian embryos
US6252133B1 (en) * 1995-08-31 2001-06-26 Roslin Institute (Edinburgh) Unactivated oocytes as cytoplast recipients of quiescent and non-quiescent cell nuclei, while maintaining correct ploidy
US6525243B1 (en) * 1995-08-31 2003-02-25 Roslin Institute Unactivated oocytes as cytoplast recipients of quiescent cell nuclei while maintaining correct ploidy
US5945577A (en) * 1997-01-10 1999-08-31 University Of Massachusetts As Represented By Its Amherst Campus Cloning using donor nuclei from proliferating somatic cells
US6215041B1 (en) * 1997-01-10 2001-04-10 University Of Mmassachusetts Cloning using donor nuclei from a non-quiesecent somatic cells
US6235969B1 (en) * 1997-01-10 2001-05-22 University Of Massachusetts Cloning pigs using donor nuclei from non-quiescent differentiated cells
US6235970B1 (en) * 1997-01-10 2001-05-22 University Of Massachusetts, Amherst Campus CICM cells and non-human mammalian embryos prepared by nuclear transfer of a proliferating differentiated cell or its nucleus
US6603059B1 (en) * 1997-03-06 2003-08-05 Infigen, Inc. Method of cloning animals
US6548741B2 (en) * 1998-10-12 2003-04-15 Geron Corporation Developmental competence for assisted reproduction and nuclear transfer in pigs

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050172347A1 (en) * 1995-08-31 2005-08-04 Campbell Keith H.S. Unactivated oocytes as cytoplast recipients for nuclear transfer
US20060064763A1 (en) * 1995-08-31 2006-03-23 Roslin Institute (Edinburgh), The Minister of Agriculture, Fisheries & Food Unactivated oocytes as cytoplast recipients for nuclear transfer
US20060156424A1 (en) * 1995-08-31 2006-07-13 Roslin Institute (Edinburgh); Minister of Agriculture, Fisheries & Food Quiescent cell populations for nuclear transfer
US7432415B2 (en) 1995-08-31 2008-10-07 Roslin Institute (Edinburgh) Unactivated oocytes as cytoplast recipients for nonprimate mammalian and pig nuclear transfer
US7514258B2 (en) 1995-08-31 2009-04-07 Roslin Institute (Edinburgh) Mammalian cultured inner cell mass cell culture using a quiescent cell as nuclear
US7524677B2 (en) 1995-08-31 2009-04-28 Rosiin Institute (Edinburgh) Mammalian cultured inner cell mass cell culture using a g1 cell as nuclear donor

Also Published As

Publication number Publication date
DE69611793T2 (en) 2001-09-20
EP1005789A3 (en) 2005-03-02
GB2318578A (en) 1998-04-29
CN100422317C (en) 2008-10-01
EP0930009A1 (en) 1999-07-21
US6147276A (en) 2000-11-14
NZ316149A (en) 1999-10-28
KR19990044148A (en) 1999-06-25
US7232938B2 (en) 2007-06-19
DE69611793D1 (en) 2001-03-22
CN1769431A (en) 2006-05-10
US7514258B2 (en) 2009-04-07
NO980845D0 (en) 1998-02-27
NZ334288A (en) 2000-06-23
GB2318578B (en) 2000-01-19
KR20060053008A (en) 2006-05-19
CA2229568A1 (en) 1997-03-06
NO980845L (en) 1998-04-29
KR100793220B1 (en) 2008-01-10
US20070028313A1 (en) 2007-02-01
KR100533476B1 (en) 2006-04-21
CN1202084A (en) 1998-12-16
HK1004938A1 (en) 1998-12-18
PL325331A1 (en) 1998-07-20
US20050010966A1 (en) 2005-01-13
CN1769432A (en) 2006-05-10
ES2155197T3 (en) 2001-05-01
SI0849990T1 (en) 2001-06-30
JP4081613B2 (en) 2008-04-30
DK0849990T3 (en) 2001-04-17
JP2006217913A (en) 2006-08-24
EP1005789A2 (en) 2000-06-07
KR20070047848A (en) 2007-05-07
WO1997007669A1 (en) 1997-03-06
US20060156424A1 (en) 2006-07-13
AU6831096A (en) 1997-03-19
EP0849990A1 (en) 1998-07-01
AU716956B2 (en) 2000-03-09
EP0849990B1 (en) 2001-02-14
GB9802915D0 (en) 1998-04-08
NZ337311A (en) 2008-05-30
SG75155A1 (en) 2000-09-19
BR9610034B1 (en) 2011-03-09
HK1090089A1 (en) 2006-12-15
US7355094B2 (en) 2008-04-08
IL123298A0 (en) 1998-09-24
AU716956C (en) 2002-03-28
BR9610034A (en) 1999-12-21
CN1230533C (en) 2005-12-07
HUP9900234A3 (en) 2000-03-28
JP2000506722A (en) 2000-06-06
ZA967390B (en) 1998-03-02
KR20050088163A (en) 2005-09-01
HUP9900234A2 (en) 1999-05-28
CZ60898A3 (en) 1998-07-15
ATE199115T1 (en) 2001-02-15
CA2229568C (en) 2011-11-22
GB9517780D0 (en) 1995-11-01

Similar Documents

Publication Publication Date Title
US7355094B2 (en) Methods of ungulate nuclear transfer
US7329796B2 (en) Serial nuclear transfer of ungulate embryos
US7304204B2 (en) Ungulates produced by nuclear transfer of G1 cells
GB2331751A (en) Quiescent cell populations for nuclear transfer
AU2003271356B9 (en) Quiescent cell population for nuclear transfer
US7361804B1 (en) Unactivated oocytes in nuclear transfer to produce ungulates
AU2005246962A1 (en) Quiescent cell population for nuclear transfer
MXPA98001646A (en) Cellular populations inactive for transfer celu

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION