WO2006071435A2 - Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals - Google Patents

Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals Download PDF

Info

Publication number
WO2006071435A2
WO2006071435A2 PCT/US2005/043224 US2005043224W WO2006071435A2 WO 2006071435 A2 WO2006071435 A2 WO 2006071435A2 US 2005043224 W US2005043224 W US 2005043224W WO 2006071435 A2 WO2006071435 A2 WO 2006071435A2
Authority
WO
WIPO (PCT)
Prior art keywords
nuclear transfer
egg
enucleated
nuclei
mannitol solution
Prior art date
Application number
PCT/US2005/043224
Other languages
French (fr)
Other versions
WO2006071435A3 (en
Inventor
Gerald Schatten
Calvin Simerly
Christopher Navara
Original Assignee
Magee-Womens Research Institute
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 Magee-Womens Research Institute filed Critical Magee-Womens Research Institute
Priority to EP05852473A priority Critical patent/EP1828376A4/en
Priority to JP2007544448A priority patent/JP2008521438A/en
Publication of WO2006071435A2 publication Critical patent/WO2006071435A2/en
Publication of WO2006071435A3 publication Critical patent/WO2006071435A3/en

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
    • A01K67/0273Cloned animals
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • 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
    • C12N15/8776Primate embryos
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0604Whole embryos; Culture medium therefor
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0608Germ cells
    • C12N5/0609Oocytes, oogonia
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/106Primate
    • 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
    • C12N2517/00Cells related to new breeds of animals
    • C12N2517/04Cells produced using nuclear transfer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Environmental Sciences (AREA)
  • Reproductive Health (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Neurosurgery (AREA)
  • Cardiology (AREA)

Abstract

The present invention is directed to various methodologies to make NT a practical procedure for animals, specifically, primates including human and non-human primates. Furthermore, the methods and molecular components provided by the present invention provide a practical means for producing embryos with desired characteristics. In a specific embodiment, the methodology of the present invention comprises introducing nuclei having desired characteristics along with one or more molecular components into an enucleated egg, thus creating a nuclear transfer construct, culturing the egg to produce a viable embryo, transferring the embryo to the oviducts of a female, and producing a cloned animal.

Description

METHODS FOR CORRECTING MITOTIC SPINDLE DEFECTS AND OPTIMIZING PREIMPLANTATION EMBRYONIC DEVELOPMENTAL RATES ASSOCIATED WITH SOMATIC CELL NUCLEAR TRANSFER IN ANIMALS
STATEMENTREGARDINGFEDERALLYSPONSOREDRESEARCH
This invention was made, at least in part, with U.S. government support under grant numbers NIH R37 HD 12913 and 2 R24 RR013632-06, awarded by NIH. The U.S. government may have certain rights in the invention. CROSS REFERENCE TO RELATED APPLICATION
The present application is a continuation-in-part of Application Serial No. 10/821,200 which claims the benefit, under 35 U.S.C. § 119, of U.S. Provisional Patent Application Serial Number 60/461,139, filed 9 April 2003, the contents of which are incorporated herein by reference. FIELD OF THE INVENTION
The present invention relates to methods for the clonal propagation of animals, including primates. The present invention also relates to methods for producing embryonic stem cells, transgenic embryonic stem cells, and immune-matched embryonic stem cells from primates, including humans. Furthermore, the present invention provides various methodologies and molecular components that may be used for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with nuclear transfer.
BACKGROUND OF THE INVENTION
Identical primates have immeasurable importance for molecular medicine, as well as implications for endangered species preservation and infertility. The lack of genetic variability among cloned animals results in a proportional increase in experimental accuracy, thereby reducing the numbers of animals needed to obtain statistically significant data, with perfect controls for drug, gene therapy, and vaccine trials, as well as diseases and disorders due to aging, environmental, or other influences. The "nature versus nurture" questions regarding the genetic versus environmental, including maternal environment or epigenetic influences on health and behavior may also be answered. Consequently, genetically identical offspring, even with differing birth dates, may be investigated (e.g., in studies such as phenotypic analysis prior to animal production; and in serial transfer of germ line cells (such as male germ cells), Brinster et al., 9 SEMIN. CELL DEV. BIOL. 401-09 (1998)), to address cellular aging beyond the life expectancy of the first offspring; and to test simultaneous retrospective (in the older twin) and prospective therapeutic protocols. Epigenetic investigations may be tested using identical embryos of the present invention implanted serially in the identical surrogate to demonstrate that, for example, low birth weight or other aspects of fetal development may have life-long consequences (Leese et al., 13 HUM. REPROD. 184-202 (1998)), the decrease in the IQ of children is related to maternal hypothyroidism during pregnancy (Haddow et al., 341 N. ENGL. J. MED. 549-55 (1999)), or immunogenetics results in uterine rejection (Gerard et al., 23 NAT. GENET. 199-202 (1999); Clark et al., 41 AM. J. REPROD. IMMUNOL. 5-22 (1999); and Hiby et al., 53 TISSUE ANTIGENS 1-13 (1999)).
The cloning of animals by adult somatic cell nuclear transfers has lead to the creation of sheep (Wilmut et al., 385 NATURE 810-13 (1997)), cattle (Kato et al., 282 SCIENCE 2095- 98 (1998)), mice (Wakayama et al., 394 NATURE 369-74 (1998)), pigs, cats, rabbits and goats (Baguisi et al., 17 NATURE BIOTECH. 456-61 (1999)). Among the most compelling scientific rationales for cloning is the production of disease models. Cloned animals as models for disease show great promise because the genetics of each clone are invariable.
Stem cell lines have been produced from human and monkey embryos (Shamblott et al., 95 PROC. NATL. ACAD. SCI. USA 13726-31 (1999) and Thomson et al., 282 SCIENCE 1145-47 (1999)). It is not yet known if stem cells from the fully outbred populations of humans or primates have the full totipotency of those from selected inbred mouse strains with invariable genetics. This can now be evaluated within the context of the present invention, for example, by producing therapeutic stem cells from one multiple, later tested in its identical sibling, and in so doing, learning if stem cells might produce cancers like teratocarcinomas.
Theoretically, somatic cell nuclear transfer (SCNT) has the potential to produce limitless identical offspring; however, genetic chimerism, fetal and neonatal death rates (Wilmut et al., 419 NATURE 583-7 (2002); Humpherys et al., 99 PROC. NATL. ACAD. SCI. USA 12889-94 (2002); Cibelli et al., 20 NAT. BiOTECHNOL. 13-4 (2002); and Kato et al., 282 SCIENCE 2095-8 (1998)), shortened telomeres (Shields et al., 399 NATURE 316-7 (1999)), and inconsistent success rates preclude its immediate usefulness. SCNT in macaques has succeeded with blastomere nuclei (Wolf et al., 60 BIOL. REPROD. 199-204 (1999)), but not yet with adult, fetal, or embryonic stem (ES) cells. These concerns notwithstanding, the contradictions and paradoxes raised by SCNT have stimulated new studies on the molecular regulation of mammalian cloning by SCNT.
Figure 1 illustrates the manipulations and developmental events that culminate in the somatic nucleus within the activated enucleated oocyte during SCNT. These steps include, but are not limited to, enucleation or metaphase-II arrested meiotic spindle removal, somatic cell selection and preparation, nuclear transfer or intracytoplasmic nuclear injection (ICNI), wound healing and drug recovery from both spindle removal and nuclear introduction, and oocyte activation. There are, however, limitations associated with these steps. For example, meiotic spindle removal in primate oocytes (non-human and human like) has unforeseen consequences on the now enucleated oocyte. Unlike oocytes from domestic species of mice, crucial microtubule motors (kinesins) and centrosome molecules (NuMA) are concentrated almost exclusively on the met-II spindle. Removal of the egg DNA along with this spindle eliminates the majority of these motors and spindle pole proteins so that the SCNT reconstituted oocyte no longer has the ability to form a functional bipolar mitotic spindle at first mitosis.
In addition, there is also a lack of fundamental scientific knowledge related to some of these steps. For instance, somatic cell preparation and selection (Wilmut et al., 385 NATURE 810-3 (1997); Wilmut et al., 419 NATURE, 583-7 (2002); and Wakayama et al., 394 NATURE 369-74 (1998)) has only been investigated in a small number of species, a comparison between electrofusion versus direct injection (Intracytoplasmic nuclear injection (ICNI)) has not been fully investigated, and wound healing after microinjection, cell fusion and 'enucleation' has yet to be investigated.
SCNT by nuclear transfer (NT; 'Dolly' approach) (Wilmut et al., 419 NATURE 583-7 (2002); Polejaeva et al., 407 NATURE 86-90 (2000); and Campbell et al., 380 NATURE 64-6 (1996)) and by ICNI (Honolulu method) (Wakayama et al., 394 NATURE 369-74 (1998) and Dominko et al., 1 CLONING 143-152 (1999)) both hold promise for propagating identical primates, but previously unanticipated biological hurdles, found only in primates, exist. Furthermore, crucial investigations regarding human embryonic stem cell potentials are investigated with non-human primates. Sets of genetically identical primate offspring would be invaluable for biomedical and behavioral investigations, yet none have been born yet. Therefore, the present invention provides reliable and effective methods for propagating identical and transgenic animals, and specifically primates. Furthermore, the present invention also provides various methodologies and molecular components that may be used for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with NT.
SUMMARY OF THE INVENTION
The present invention is directed to various methodologies to make NT a practical procedure for animals, and specifically primates. Furthermore, the methods and molecular components provided by the present invention provide a practical means for producing embryos with desired characteristics. In one embodiment, the methodology of the present invention may include introducing nuclei into an extrusion-enucleated egg, thus creating a nuclear transfer construct, culturing the nuclear transfer construct to produce a viable embryo, transferring the embryo to the oviducts of a female, and producing a cloned animal.
In one embodiment of the present invention, the method may comprise steps of introducing nuclei along with one or more molecular components into an extrusion- enucleated egg, thus creating a nuclear transfer construct; culturing said nuclear transfer construct to produce a viable embryo; transferring said embryo to the oviducts of a female; and producing a cloned animal. In one embodiment of the present method, the enucleated egg may comprise a a cumulus-free oocyte. In another particular embodiment, the methods may utilize an enucleated egg that is enucleated pre-metaphase II. In another particular embodiment of the present invention, the methods may utilize an enucleated egg that is enucleated just prior to metaphase II arrest. In another embodiment of the methods of the present invention, extrusion may comprise holding an egg with a holding micropipette; partially dissecting the zonal pellucida of the egg with a needle by making a slit near the first polar body of said egg; extruding the first polar body and adjacent cytoplasm containing the meiotic spindle, ranging from about telophase-I to about pro-metaphase-II, by squeezing the needle. In another embodiment of the present invention, the methods may utilize a holding micropipette that has an about 110 μm inner diameter. In another particular embodiment, the methods of the present invention may utilize a glass needle to extrude the egg's nucleus.
In one embodiment of the methods of the present invention, the egg may be enucleated in Hepes-buffered TALP supplemented with BSA and cytochalasin B. In another embodiment of the methods of the present invention, the egg may be enucleated in Hepes- buffered TALP supplemented with about 0.3% BSA and about 7.5 μg/ml cytochalasin B. The transferred nuclei of the methods of the different invention may come from different sources. For example, and without limitation, in one embodiment of the methods of the present invention, the nuclei may be derived from a somatic cell nuclear donor source. In another particular embodiment of the methods of the present invention, the nuclei may be derived from a somatic cell nuclear donor source that may include dissociated cumulus cells. In another particular embodiment of the methods of the present invention, the dissociated cumulus cells may be autologous. In another particular embodiment, the dissociated cumulus cells may be heterologous. In another particular embodiment of the methods of the present invention, the cumuls cells may be autologous and heterologous. In another particular embodiment of the methods of the present invention, the nuclei may be derived from primary rhesus fibroblast cell lines. In another particular embodiment, the methods of the present invention may utilize nuclei that may be derived from donor blastomeres.
In a particular embodiment of the methods of the present invention, the nuclei may be transferred into the perivitelline space of an enucleated egg to create a nuclear transfer construct. In a particular embodiment, the nuclear transfer constructs are equilibrated with mannitol solution. In another particular embodiment of the present invention, the methods may utilize mannitol solution comprising about 0.3 M mannitol solution containing about 0.5 niM Hepes, about 0.1 mM CaC12, and about 0.1 mM MgC12. In another particular embodiment of the present invention, the methods may equilibrate the nuclear transfer constructs with mannitol solution for about 4 minutes. In another particular embodiment of the methods of the present invention, the nuclear transfer constructs may be transferred to a chamber containing an electrode overlaid with the mannitol solution after the constructs' equilibration with mannitol solution. In another particular embodiment, the chamber may contain more than one electrode. In another particular embodiment, the methods of the present invention may utilize a chamber that may include 2 electrodes. In one embodiment of the present invention, the methods may fuse the nuclei and egg with two DC pulses. In another particular embodiment, the methods of the present invention may fuse the nuclei and the egg with DC pulses constitute of about 2.7 kK/cm. In another particular embodiment, the DC pulses may be of a duration of about 15 μs. In one embodiment of the methods of the present invention, the nuclear transfer construct may be developed in culture media. In one embodiment the culture media may include of Gl, G2, and modified synthetic oviductal fluid (mSOF). In another particular embodiment, the methods of the present invention may develop the nuclear transfer construct in the culture media sequentially. In another particular embodiment, the nuclear transfer construct may be developed in Gl for about 48 hours after nuclear transfer, then developed in G2 media for about another 48 hours followed by transfer to mSOF around the morula stage until the nuclear transfer construct reaches the blastocyst stage. In another particular embodiment of the methods of the present invention, the mSOF media may further comprise fructose.
In a specific embodiment, nuclei with desired characteristics may be obtained by selection or by design and transferred into eggs, for example, enucleated eggs. In a particular embodiment, normally occurring nuclei may be selected for genetic compatibility or complementarity to a host or may be derived or engineered from donors with desirable characteristics. In another embodiment of the present invention, the desired characteristics may be linked to a specific disease or disorder. In particular, the disease or disorder may comprise cardiovascular disease, neurological disease, reproductive disorder, cancer, eye disease, endocrine disorder, pulmonary disease, metabolic disorder, autoimmune disorder, and aging. Selected nuclei may be introduced into eggs along with molecular components comprising centrosomal components normally present in sperm centrosomes. In another embodiment, the molecular components comprise mitotic motor proteins and centrosome proteins, such as kinesins (e.g., HSET) and NuMA, respectively.
In particular, the methods of the present invention may comprise double nuclear transfer; meiotic spindle collapse, maternal DNA removal, and recovery; pronuclear removal after NT and fertilization or artificial activation; and cytoplasmic transfer or ooplasm supplementation.
In another embodiment of the present invention, the animal may be a mammal, bird, reptile, amphibian, or fish. In another aspect of this method, the animal may be a non-human primate, and in particular, a monkey. In an alternate aspect of this method, the animal may be a primate, and in particular, a human. In a particular embodiment of the present invention, the animal may be transgenic. In another embodiment, the present invention provides cloned animals produced by the methods of the present invention.
In a specific embodiment of the present invention, preimplantation genetic diagnosis may be performed on a blastomere isolated from the embryo prior to transfer to the oviduct of a female surrogate. The methods used for this preimplantation genetic diagnosis include polymerase chain reaction (PCR), fluorescence in situ hybridization (FISH), single-strand conformational polymorphism (SSCP), restriction fragment length polymorphism (RFLP), primed in situ labeling (PRINS), comparative genomic hybridization (CGH), single cell gel electrophoresis (COMET) analysis, heteroduplex analysis, Southern analysis and denatured gradient gel electrophoresis (DGGE) analysis.
Also within the scope of the present invention is the production of embryos and stem cells, such as embryonic stem cells and transgenic embryonic stem cells, using the methods of the present invention. In a specific embodiment, SCNT embryos are used to produce clonal offspring and the isolated blastomeres are used to produce an embryonic stem cell line. In a further embodiment, the SCNT embryos are transgenic, and these SCNT transgenic embryos are used to produce clonal transgenic offspring and the isolated transgenic blastomeres are used to produce transgenic embryonic stem cell lines.
The present invention also relates to methods of producing embryonic stem cells whereby blastomeres are dissociated from embryos and these cells are then cultured to produce stem cell lines. In a specific embodiment, the methods described herein are used to produce primate embryonic stem cells. In another aspect of the invention, the methods described herein are used to produce transgenic embryonic stem cells including, for example, transgenic primate embryonic stem cells.
The present invention is also directed to embryonic stem cells produced by the methods described herein. In a particular embodiment, the embryonic stem cells are primate embryonic stem cells. In a further embodiment, the embryonic stem cells are transgenic including, for example, transgenic primate embryonic stem cells. In yet another embodiment, the transgenic embryonic stem cells are human transgenic embryonic stem cells.
The present invention also relates to methods for preimplantation genetic diagnosis of an embryo. In a specific embodiment, blastomeres are dissociated from an embryo and genetic analysis is performed on a single blastomere. In a further embodiment of the present invention, the remaining blastomeres are cultured to an embryonic stage and subsequently implanted in a female surrogate. The methods used for the genetic analysis of the blastomere include PCR, FISH, SSCP, RFLP, PRINS, CGH, COMET analysis, heteroduplex analysis, Southern analysis, and DGGE analysis.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 provides a schematic illustration of the manipulations and events that occur during SCNT. The steps include enucleation or metaphase-II arrested meiotic spindle removal, somatic cell selection and preparation, nuclear transfer (NT) or intracytoplasmic nuclear injection (ICNI), wound healing and drug recovery from both spindle removal and nuclear introduction, and oocyte activation.
Figures 2A-2G illustrate that faulty mitotic spindles produce aneuploid embryos after primate NT. Figure 2A illustrates a defective NT mitotic spindle with misaligned chromosomes centrosomal NuMA at meiosis. Figure 2B illustrates a defective NT mitotic spindle with misaligned chromosomes centrosomal NuMA at mitosis. Figure 2C illustrates that a defective NT mitotic spindle with misaligned chromosome centrosomal NuMA does not occur at NT mitosis. Figure 2D illustrates that centrosomal kinesin HSET is also missing after NT. Figure 2E illustrates that centromeric Eg5 is not missing after NT. Figure 2F illustrates that bipolar mitotic spindles are with aligned chromosomes and centrosomal NuMA after NT into fertilized eggs. Figure 2G provides DNA microtubule, NuMA, and kinesin imagining.
Figures 3A-3R provide a schematic illustration of manipulations and events that occur during therapeutic cloning. These steps, which are described herein, generally include oocyte collection, enucleation, nuclear transfer, activation, cell division and differentiation, and transfer to the patient.
Figures 4A-4L show SCNT NHP embryo preimplantation development in vitro. Figure 4A shows a Spindle View™ image of the just formed metaphase-II spindle (arrow) in a living NHP oocyte. The first polar body (Pb) is visible just above the bipolar spindle structure. Figure 4B shows a karyoplast formed from 'squish' enucleation of an oocyte just after polar body extrusion. The telophase-I spindle is visible after it is immunolabeled with HSET antibody (green; inset: microtubules, red) and Hoechst DNA stain of the meiotic chromosomes (blue). Figure 4C shows a SCNT construct from 8 hours post activation following nuclear transfer by electrofusion. This Figure shows the single nucleus in the activated cytoplasm. The inset of Figure 4C is a representation of a normal karyotype from the donor rhesus fibroblast cell line used for somatic cell nuclear transfer (SCNT). Figures 4D-4K show the in vitro development of SCNT embryos through the cleavage stages: two- cell (D), three-cell (E), eight-cell (F; arrow: slight fragmentation), 16 cell (G), the compacting morula stage (H), early blastocyst (I; arrow: early blastocoel), expanded blastocyst (J), and the hatched blastocyst stage (K). Figure 4L shows NHP ES cells derived from a NT x ICSI fertilized chimeric blastocyst, as imaged by HMC optics 4 days post outgrowth on nonhuman primate embryonic feeder (nhpEF) cells. All numbers in Figure 4 represent time post-activation except for Figure 4L which represents days post outgrowth. The bars of Figure 4 represent 20μm.
Figures 5A-5F show abnormal preimplantation development of ECNT-derived NHP embryos. Figure 5A shows a first mitotic telophase clone (green) showing atypical chromosome segregation (blue) at the end of first mitosis. Arrow points to a lagging chromosome (blue) located within the interzonal microtubules (green). Figures 5B-F show abnormal chromosome segregation (blue) and microtubule organization (green) in ECNT cloned embryos that is apparent at the 2-cell (B), 4-cell (C), 6-cell (D), and 8-cell stages (E), where most of embryonic development arrests. Figure 5F shows control 8-cell stage parthenogenote that demonstrate normal chromosome (blue) and interphase microtubule patterns (green). All images are double-labeled for microtubules (green) and DNA (blue). Pb stands for polar body while each bar represents 20 μm.
Figures 6A-6J show abnormal microtubules patterns after nuclear transfer in NHP, but not bovine, constructs. Figures 6A and 6B show disarrayed microtubules (green) assembled near the transferred somatic cell nucleus (blue) which indicates a dysfunctional somatic centrosome following intracytoplasmic nuclear injection [ICNI] and activation by either sperm factor or ionomycin/DMAP. Figure 6C shows cortical microtubule patterns (green) similar to parthenogenetically activated oocytes observed after somatic cell nuclear transfer (blue) and activation. Figure 6D shows a NHP NT construct derived by embryonic nuclear transfer using a dissociated 16-cell stage rhesus blastomere. Multiple microtubule organizing centers (green, arrows) within the cytoplasm are detected distal to the transferred blastomere nucleus (blue). Figure 6E shows NHP NT construct derived by the transfer of a male pronucleus (MPn, blue) into an enucleated oocyte. Microtubules (green) are tightly focused at the transferred nucleus and radiate into the cytoplasm. Again, Pb stands for polar body, while FPn stands for female pronucleus. Figure 6F shows DNA synthesis onset in the transferred somatic cell nucleus (sc, blue) as well as male [MPN, blue] and female [FPN, blue] pronuclei as detected by BrDU incorporation (green) 20 hrs post ICSI. Neither the first nor second polar bodies [Pb] incorporate BrDU. The inset of Figure 6F shows microtubules (red) and DNA (blue). Figures 6G-6H show tightly focused microtubule arrays (green) emanating from the transferred nucleus in activated bovine enucleated cytoplasts following either somatic cell or embryonic cell nuclear transfer. Figure 61 shows normal anastral, bipolar spindles (green) with aligned chromosomes assembled at metaphase (blue) following embryonic nuclear transfer. Similar mitotic spindle morphologies were observed after somatic cell nuclear transfer. Figure 6J shows a focused microtubule array (green) from a rhesus fibroblast cell (blue) transferred into a bovine enucleated oocyte. All images are double-labeled for microtubules (green) and DNA (blue) except for Figure 6F which is triple labeled for BrDU (green), microtubules (red) and DNA (blue). Bars represent lOμm. Figures 7A-7E show that microtubule patterns are normal in NHP androgenotes.
Figures 7 A and 7B show that a mature spermatozoa (blue) microinjected into an enucleated rhesus oocyte assembles tightly focused microtubule arrays (green) from the sperm centrioles (arrowhead: sperm axoneme) that extend into the cytoplasm within 8 hrs post ICSI. Figure 7C shows centrosome duplication where splitting and microtubule assembly (green) is observed on opposite sides of the male pronucleus by 20 hours post-ICSI. Figure 7D shows a bipolar spindle (green) with small astral arrays (green, arrows) at the spindle poles assembled at metaphase (blue) in androgenotes. The arrowhead shows the incorporated sperm axoneme. Figure 7E show a 2-cell stage androgenote demonstrating normal DNA segregation (blue) and microtubule assembly (green) near the daughter nuclei following cell division. All images are double-labeled for microtubules (green) and DNA (blue). Bars represent 20 μm.
Figures 8A-8K show that dysfunctional somatic cell centrosomes and microtubule- based molecular motors are evident in mitotic metaphase NHP constructs. Figures 8 A and 8B show a mitotic metaphase ECNT construct with tripolar spindles (green), abnormal centrosome localization (arrows) at the poles, and misaligned chromosomes at the equator (blue). Figure 8C shows a first mitotic metaphase SCNT clone with poor bipolar spindle morphology (green), no discernible somatic cell centrosome at the spindle poles, and misaligned chromosomes at the equator (blue). Figures 8D and 8F show NuMA detection in interphase and mitotic NT constructs. In Figure 8D, NuMA (green) is detected in the ECNT reconstructed nucleus at late interphase. The inset of Figure 8D shows that random disarrayed microtubule patterns (red) and DNA (blue) are observed in this ECNT clone. Similar observations were observed. in interphase SCNT constructs. Figure 8E shows a SCNT construct at first mitotic metaphase showing a multipolar spindle (red) with misaligned chromosomes (blue) and diminished NuMA detection at the poles (green). Figure 8F shows a first mitotic metaphase spindle produced from activation of a metaphase-II spindle intact oocyte after SCNT [SCNT + Met-II]. Four misplaced centrosomes (arrows) are present within the tetrapolar spindle (red) as the chromosomes align at the equator (blue). NuMA (green) is strongly detected at the centrosomes and four spindle poles. Figure 8G shows that the minus-end directed kinesin HSET (green) is not detected in SCNT first mitotic constructs. The inset of Figure 8G shows spindle microtubules (red) and misaligned DNA (blue). Figure 8H shows a first mitotic metaphase spindle in a SCNT + Met-II intact construct. HSET is strongly detected at the metaphase spindle poles (green). The inset of Figure 8H shows spindle microtubules (red) and DNA (blue). Figure 81 shows the plus-end directed kinesin Eg5 detection in a first mitotic SCNT spindle. The multipolar metaphase spindle (red)) shows Eg5 present at the centromere region on the misaligned chromosomes (blue). Figures 8J-8K show mitotic metaphase and telophase spindles in control parthenogenetic embryos. Eg5 (green) is detected at aligned chromosomes (blue) on bipolar metaphase spindles (red), but translocates to interzonal microtubules (K: red) and the developing midbody apparatus by telophase (K: blue). Figures 8A-8C are double-labeled images for microtubules (green) and DNA (blue). All other images are tripled-labeled for NuMA (Figures 8D-8F), HSET kinesin (Figures 8G and 8H) or Eg5 kinesin (Figures 8I-8K), microtubules (red), and DNA (blue). Bars represent 10 μm.
Figures 9A-9 J show that the minus-end directed kinesin HSET assembles exclusively at the second meiotic spindle in NHP' s and not in taxol-induced cytoplasmic microtubules. Figures 9A-9C show microtubule patterns (green) observed in rhesus cytoplast after enucleation (Figure 9A) or following artificial activation at 24 (Figure 9B) or at 48 hrs (Figure 9C) post-ionomycin/DMAP. Following enucleation, no assembled cytoplasmic 1 microtubules (Figure 9A: green) are observed. After activation, abundant disarrayed microtubules (Figure 9B: green) assemble in the DNA-less cytoplast. Rarely, a microtubule structure resembling a bipolar spindle (Figure 9C: green) assembles in the cytoplasm. The insets of Figures 9A-9C show DNA imaging confirming successful removal of the SCC. Figure 9D shows microtubules (red, inset), HSET (green) and DNA (blue) imaging of the intact meiotic spindle and chromosomes in a karyoplast following 'squish' enucleation. Figures 9E and 9F show cytoplasmic HSET (green) detection in NHP cumulus (In Figure 9E green; blue, DNA) and rhesus fibroblast cells (Figure 9F green; red, microtubules; blue, DNA) shows that some somatic cell HSET may be transferred during NT. Figure 9G shows a mature oocyte treated for 30 minutes with lOμM paclitaxel demonstrating HSET (green) localization at the spindle pole microtubules (red), though not at assembled cytoplasmic microtubule bundles (arrows). Second meiotic chromosomes, are blue. Figures 9H-9J show a rhesus enucleated cytoplast treated for 30 minutes with lOμM paclitaxel prior to fixation and detection of DNA (H, blue), microtubules (I, red), and HSET (J, green). The assembled cytoplasmic microtubule bundles are not labeled by HSET antibody. Figures 9A-9C and 9E are double-labeled images for microtubules (green) and DNA (blue). Figures 9D, 9F, and 9G are triple-labeled images for HSET (green), microtubules (red) and DNA (blue). Figures 9H- 9J are single-labeled images for DNA (blue), microtubules (red), and HSET (green). Bars represent lOμm. Figures 10A-10H show that the spindle pole matrix protein NuMA is not restricted to the second meiotic spindle but also resides in the cytoplasm of NHP oocytes after SCC enucleation. Figure 1OA shows NuMA (green), microtubules (red) and DNA (blue) detection of the intact second meiotic spindle removed by 'squish' enucleation. Figure 1OB shows the enucleated cytoplast, formed after removal of the SCC by 'squish' enucleation, and no NuMA (green) or microtubule assembly (top inset: red; bottom inset: DNA, blue) in the cytoplasm. Figure 1OC shows a mature NHP oocyte treated for 20 minutes with lOμM paclitaxel which demonstrates NuMA (green) accumulation within the second meiotic spindle (lower arrow; red, microtubules; blue, DNA) and the cytoplasmic microtubule bundles (upper arrow; red, microtubules; blue, DNA). Figures 1OD and 1OE show NuMA detection in the somatic cell nuclei of cumulus (Figure 10D: green) and rhesus fibroblast cells (Figure 1OE: green; red, microtubules). Figure 1OF shows a 'FertClone' failure derived from an oocyte that failed nuclear transfer of the fibroblast cell by electrofusion (arrowhead), but was successfully activated by intracytoplasmic sperm injection (ICSI). NuMA (green) is strongly detected in the decondensed male pronucleus (MPn), but diminished in the unsuccessful fibroblast cell (arrowhead) attached at the oocyte surface. In the upper inset of Figure 1OF, red depicts microtubules while blue depicts DNA. In the lower inset of Figure 1OF, the sperm centrosome organizes a microtubule astral array (arrow) near the decondensed male pronucleus. Figure 1OG shows a 'Fert-Clone' failure produced by SCNT into an intact second meiotic metaphase-II arrested oocyte that unsuccessfully activated following ICSI (arrow). NuMA (green) is found at both poles on the intact metaphase-II spindle (red) with aligned chromosomes (blue) as well as at the base of the condensed sperm head (blue, arrow). However, NuMA (green) is missing from the disorganized multipolar NT spindle microtubules (red) assembling around the scattered somatic cell chromosomes (blue). Figure 1OH shows ECNT into a metaphase-II intact oocyte that failed subsequent activation by sperm factor microinjection. NuMA (green) is detected at the poles in both the NT spindle (lower arrow; red, microtubules; blue, DNA) and the intact second meiotic metaphase spindle (upper arrow; red, microtubules; blue, DNA). All Figure 10 images are triple-labeled for NuMA (green), microtubules (red) and DNA (blue) except Figure 1OD which is single labeled for NuMA (green) and Figure 1OE which is double-labeled for microtubules (red) and NuMA (green). Bars represent lOμm, except for the bar in Figure 1OD which represents lμm.
Figure 11 depicts centrosome transmission during primate nuclear transfer (right) and fertilization (left). NT begins with 'squish' enucleation (Top right), the removal of the unfertilized oocyte's pre-metaphase-II meiotic spindle-chromosome complex (SCC), leaving some NuMA (Green crosslinker) and HSET (Red pacman) molecular motor protein remaining in the ooplasm. Better retention of vital proteins (i.e., myosin-II, fodrin, Wave-1; actin filaments: crosshatches) in the meiotic spindle cortex also is anticipated following 'squish' enucleation. In the right panel of Figure 11, enucleation of the pre-metaphase-II SCC removes less of the minus-end directed spindle proteins NuMA (Green crosslinker) and HSET motors (Red pacman). Referring to about the middle of the right panel of Figure 11, nuclear transfer by electrofusion (yellow lightening bolt) introduces an embryonic or somatic nucleus and centrioles (red orthogonal cylinders) containing γtubulin and pericentral (red lattice) into the enucleated cytoplast, as well as providing the simultaneous activating stimulus to initiate development. The bottom right panel shows that enucleation by 'squish' extrusion along with simultaneous fusion/activation results in more organized bipolar first mitotic spindles. Figure 11 also depicts that NT-mitotic spindles display mostly aligned chromosome pairs (blue) with Eg5 at their centromere/kinetochore regions (Yellow pacman). Microtubules assemble (Green) into organized spindles with both NuMA (Green crosslinker) and HSET (Red pacman) present at their spindle poles.
The left panel of Figure 11 shows that sperm entry, either by IVF or ICSI (shown), activates the egg's metabolism and contributes the paternal haploid genome to the now fertilized zygote. The typical meiotic spindle arrested at second metaphase (Blue chromosomes) is unusual since it lacks centrioles at the poles. The plus-end directed kinesin motor, Eg5, concentrated at the centromeres (Yellow pacman), anchors at the growing-end (+) of the polarized microtubules (Green). Centrosome molecules NuMA (Green crosslinker) and HSET (Red pacman), responsible for meiotic spindle organization, are concentrated at the converging-microtubule (-) ends and would be removed during enucleation at this stage. The middle of the left panel of Figure 11 shows that the fertilizing sperm contributes the centriole pair (red orthogonal cylinders) containing paternal γ-tubulin and pericentrin (Red lattice). This sperm centrosome complex recruits maternal γ-tubulin from which sperm aster microtubules assemble (Green). The bottom of the left panel of Figure 11 shows first mitotic spindle assembly after fertilization. The sperm centrosome duplicates during first interphase, with the sperm tail-centriole complex visible at one pole of the bipolar, anastral spindle. The bipolar mitotic spindle contains aligned chromosomes (Blue) with Eg5 at each kinetochore pair (Yellow pacman). NuMA (Green crosslinker) and HSET (Red pacman) are found at the spindle poles along with the centrioles and γ-tubulin/pericentrin (Red lattice). The other spindle pole (demarked by a ? mark) is either organized without a centriole pair or contains centrioles of unknown derivation.
DETAILED DESCRIPTION OF THE INVENTION It is understood that the present invention is not limited to the particular methodology, protocols, and reagents, etc. described herein, as these may vary. It is also to be understood that the terminology used herein is used for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention. It must be noted that as used herein and in the appended embodiments, the singular forms "a," "an," and "the" include plural reference unless the context clearly dictates otherwise.
Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Preferred methods, devices, and materials are described, although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention. All references cited herein are incorporated by reference herein in their entirety.
DEFINITIONS
For convenience, the meaning of certain terms and phrases employed in the specification, examples, and appended embodiments are provided below. The term "animal" includes all vertebrate animals such as mammals (e.g., rodents, mice and rats), primates (e.g., monkeys, apes, and humans), sheep, dogs, rabbits, cows, pigs, amphibians, reptiles, fish, and birds. It also includes an individual animal in all stages of development, including embryonic and fetal stages.
The term "primate" as used herein refers to any animal in the group of mammals, which includes, but is not limited to, monkeys, apes, and humans.
The term "totipotent" as used herein refers to a cell that gives rise to all of the cells in a developing cell mass, such as an embryo, fetus, and animal. In specific embodiments, the term "totipotent" also refers to a cell that gives rise to all of the cells in an animal. A totipotent cell can give rise to all of the cells of a developing cell mass when it is utilized in a procedure for creating an embryo from one or more nuclear transfer steps. An animal may be an animal that functions ex utero. An animal can exist, for example, as a live born animal. Totipotent cells may also be used to generate incomplete animals such as those useful for organ harvesting, e.g., having genetic modifications to eliminate growth of a head, or other organ, such as by manipulation of a homeotic gene. The term "totipotent" as used herein is to be distinguished from the term
"pluripotent." The latter term refers to a cell that differentiates into a sub-population of cells within a developing cell mass, but is a cell that may not give rise to all of the cells in that developing cell mass. Thus, the term "pluripotent" can refer to a cell that cannot give rise to all of the cells in a live born animal. The term "totipotent" as used herein is also to be distinguished from the term
"chimeric" or "chimera." The latter term refers to a developing cell mass that comprises a sub-group of cells harboring nuclear DNA with a significantly different nucleotide base sequence than the nuclear DNA of other cells in that cell mass. The developing cell mass can, for example, exist as an embryo, fetus, and/or animal. The term "embryonic stem cell" as used herein includes pluripotent cells isolated from an embryo that may be maintained, for example, in in vitro cell culture. Embryonic stem cells may be cultured with or without feeder cells. Embryonic stem cells can be established from embryonic cells isolated from embryos at any stage of development, including blastocyst stage embryos and pre-blastocyst stage embryos. Embryonic stem cells and their uses are well known to a person of skill in the art. See, e.g., U.S. Patent No. 6,011,197 and WO 97/37009, entitled "Cultured Inner Cell Mass Cell-Lines Derived from Ungulate Embryos," Stice and Golueke, both of which are incorporated herein by reference in their entireties, including all figures, tables, and drawings, and Yang & Anderson, 38 THERIOGENOL. 315-35 (1992).
For the purposes of the present invention, the term "embryo" or "embryonic" as used herein includes a developing cell mass that has not implanted into the uterine membrane of a maternal host. Hence, the term "embryo" as used herein can refer to a fertilized oocyte, a cybrid, a pre-blastocyst stage developing cell mass, and/or any other developing cell mass that is at a stage of development prior to implantation into the uterine membrane of a maternal host. Embryos of the invention may not display a genital ridge. Hence, an "embryonic cell" is isolated from and/or has arisen from an embryo.
An embryo can represent multiple stages of cell development. For example, a one cell embryo can be referred to as a zygote, a solid spherical mass of cells resulting from a cleaved embryo can be referred to as a morula, and an embryo having a blastocoel can be referred to as a blastocyst.
The term "fetus" as used herein refers to a developing cell mass that has implanted into the uterine membrane of a maternal host. A fetus can include such defining features as a genital ridge, for example. A genital ridge is a feature easily identified by a person of ordinary skill in the art, and is a recognizable feature in fetuses of most animal species. The term "fetal cell" as used herein can refer to any cell isolated from and/or arisen from a fetus or derived from a fetus. The term "non-fetal cell" is a cell that is not derived or isolated from a fetus. The term "inner cell mass" as used herein refers to the cells that gives rise to the embryo proper. The cells that line the outside of a blastocyst are referred to as the trophoblast of the embryo. The methods for isolating inner cell mass cells from an embryo are well known to a person of ordinary skill in the art. See, Sims & First, 91 PROC. NATL. ACAD. SCI. USA 6143-47 (1994) and Keefer et al., 38 MOL. REPROD. DEV. 264-268 (1994). The term "pre-blastocyst" is well known in the art.
A "transgenic embryo" refers to an embryo in which one or more cells contain heterologous nucleic acid introduced by way of human intervention. The transgene may be introduced into the cell, directly or indirectly, by introduction into a precursor of the cell, by way of deliberate genetic manipulation, or by infection with a recombinant virus. In the transgenic embryos described herein, the transgene causes cells to express a structural gene of interest. However, transgenic embryos in which the transgene is silent are also included.
The term "transgenic cell" refers to a cell containing a transgene. The term "germ cell line transgenic animal" refers to a transgenic animal in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration of genetic information, they are transgenic animals as well. The term "gene" refers to a DNA sequence that comprises control and coding sequences necessary for the production of a polypeptide or precursor. The polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence so long as the desired enzymatic activity is retained.
The term "transgene" broadly refers to any nucleic acid that is introduced into the genome of an animal, including but not limited to genes or DNA having sequences which are perhaps not normally present in the genome, genes which are present, but not normally transcribed and translated ("expressed") in a given genome, or any other gene or DNA which one desires to introduce into the genome. This may include genes which may be normally present in the nontransgenic genome but which one desires to have altered in expression, or which one desires to introduce in an altered or variant form. The transgene may be specifically targeted to a defined genetic locus, may be randomly integrated within a chromosome, or it may be extrachromosomally replicating DNA. A transgene may include one or more transcriptional regulatory sequences and any other nucleic acid, such as introns, that may be necessary for optimal expression of a selected nucleic acid. A transgene can be coding or non-coding sequences, or a combination thereof. A transgene may comprise a regulatory element that is capable of driving the expression of one or more transgenes under appropriate conditions.
The phrase "a structural gene of interest" refers to a structural gene, which expresses a biologically active protein of interest or an antisense RNA, for example. The structural gene may be derived in whole or in part from any source known to the art, including a plant, a fungus, an animal, a bacterial genome or episome, eukaryotic, nuclear or plasmid DNA, cDNA, viral DNA, or chemically synthesized DNA. The structural gene sequence may encode a polypeptide, for example, a receptor, enzyme, cytokine, hormone, growth factor, immunoglobulin, cell cycle protein, cell signaling protein, membrane protein, cytoskeletal protein, or reporter protein (e.g., green fluorescent protein (GFP), β-galactosidase, luciferase). In addition, the structural gene may be a gene linked to a specific disease or disorder such as a cardiovascular disease, neurological disease, reproductive disorder, cancer, eye disease, endocrine disorder, pulmonary disease, metabolic disorder, autoimmune disorder, and aging.
A structural gene may contain one or more modifications in either the coding or the untranslated regions which could affect the biological activity or the chemical structure of the expression product, the rate of expression, or the manner of expression control. Such modifications include, but are not limited to, mutations, insertions, deletions, and substitutions of one or more nucleotides. The structural gene may constitute an uninterrupted coding sequence or it may include one or more introns, bound by the appropriate splice junctions. The structural gene may also encode a fusion protein.
Primates identical in both nuclear and cytoplasmic components represent ideal scientific models, for example, for preclinical investigations on the genetic and epigenetic basis of diseases. Here, the present invention relates to producing genetically identical primates as twin and higher-order multiples by using SCNT. Further, the present invention contemplates several methods for correcting dysfunctional reproductive potential in human and non-human primates and therapeutic value of cells and tissue derived from embryos after application of NT technology. For NT to be effective, the introduced diploid nucleus from a somatic or embryonic nucleus should be capable of condensing and aligning their duplicated chromosomes on a functional bipolar spindle apparatus at first mitosis. This may be followed by accurate segregation of the aligned chromosomes at the end of first division. The assembly of a functional bipolar spindle is, in turn, reliant on the two critical events: (i) the cell's microtubule organizing center (i.e., the somatic or embryonic centrosome) introduced during nuclear transfer which nucleates the spindle microtubules after nuclear envelope breakdown; and (ii) the action of a set of structural components (i.e., including nuclear mitotic apparatus protein (e.g., NuMA) and molecular motor proteins (including kinesin motor proteins)), which are largely contributed by the egg cell, and which crosslink, organize and shape the bipolar spindle apparatus for aligning and segregating the duplicated chromosomes. Previously, there has been no evaluation of the centrosomes or structural/molecular motor proteins role in bipolar spindle assembly after nuclear transfer. The present invention illustrates that dysfunctional centrosomes as well as missing NuMA and HSET kinesin result in mitotic multipolar spindles with misaligned chromosomes and aneuploid embryos after nuclear transfer.
The following embodiments of the present invention relate to various techniques for correcting mitotic spindle defects associated with NT. The methodologies provided by the present invention are capable of evaluating mechanisms for potential nuclear transfer failures related to first mitotic errors, previously elusive of efficient detection.
There is a direct correlation between reproduction and the ability to assemble bipolar mitotic spindles, which are responsible for accurately and faithfully segregating the duplicated chromosomes. Generally, in primates, the sperm contributes the centrioles, which are critical to the assembly of a functional centrosome. Following centrosome assembly, the centrosome participates in the assembly of the first mitotic spindle microtubules. The oocyte, in contrast, contributes various motor proteins, such as members of the kinesin superfamily and dynein, which coalesce on the mitotic spindle microtubules. The function of the various motor proteins is to participate in and maintain the assembly of the bipolar mitotic spindle. The mitotic spindles are essential to the production of viable human and non-human primate embryos. It was unanticipated that spindle organization and accurate segregation of chromosomes would depend on these molecules. The necessity for these components is demonstrated by the non-viability of embryos prepared by NT that lack structural or motile molecules from the sperm centrosome. Therefore, spindle-organizing principles that are present in sperm may be required to produce useful success rates of NT and the practical production of embryos intended for producing cells, tissues or animals with selected characteristics.
The present invention contemplates that the introduction of centrosomal components may correct mitotic spindle defects associated with NT. Furthermore, the present invention provides some of the key components needed for the correction of mitotic spindle defects. In particular, these components may include, but are not limited to, NuMA and HSET kinesin.
The present invention also contemplates various methodologies that would make NT a practical procedure for animals, and specifically human or non-human primates. In particular, nuclei with desired characteristics would be obtained by selection or by design and transferred into eggs. Normally occurring nuclei may be selected for genetic compatibility or complementarity to a host or may be derived or engineered from donors with desirable characteristics. Selected nuclei would be introduced into eggs along with components normally present in sperm centrosomes. The addition of centrosomal components may be necessary for the production of viable embryos. The utility of these methods and molecules provided by the present invention creates a practical means for producing embryos with desired characteristics.
A specific embodiment of the present invention relates to the pronuclear removal after SCNT and fertilization (ICS/NI-2PN). Figures 2A-2G demonstrate the feasibility and benefit of pronuclear removal after SCNT and fertilization (ICS/NI-2PN). ICNI without prior oocyte enucleation is followed by fertilization. The somatic cell may be introduced distal from the first polar body allowing a geographical separation between the female pronucleus and the diploid nucleus. The sperm may be identified either by prelabeling its mitochondria with, for example, the vital dye MitoTracker or by imaging the incorporated sperm tail. At first interphase, the two pronuclei are extracted and the reprogrammed and remodeled somatic nucleus resides in the sperm-activated oocyte with the full complement of ooplasmic proteins restored after second meiotic completion. For a discussion of nuclear programming, see generally Wakayama et al., 5(3) CLONING AND STEM CELLS 181-89 (2003); Dinnyes et al., 4(1) CLONING AND STEM CELLS 81-90 (2002); and Jaenisch et al., 4(4) CLONING AND STEM CELLS 389-96 (2002).
Additionally, the present invention also contemplates a second NT so that the somatic nucleus may be reprogrammed and remodeled as during NT (double NT). However, the nucleus may be transferred the next day into another egg that had been fertilized by intracytoplasmic sperm injection (ICSI) previously. The male and female pronuclei (sperm and egg nuclei, respectively) of the zygote may be removed by micromanipulation. The second nuclear transfer, from the first interphase NT into the now enucleated zygote, inserts the reprogrammed somatic diploid nucleus into an interphase cytoplasm that has been activated by the sperm and contains all the ooplasmic constituents previously sequestered on the meiotic spindle. Normally, the spindle-associated motors are returned to the ooplasm after second polar body formation, and with the double NT strategy, they are in full complement. Double NT affords several advantages, including its successful application during SCNT in pigs. However, it requires twice the number of eggs and many more demanding intricate procedures, including pronuclear extraction coupled by interphase nuclear transfer.
Another embodiment of the present invention contemplates meiotic spindle collapse using reversible microtubule disassembly with either nocodazole or cold to reduce or eliminate the spindle microtubules. Dynamic DNA imaging identifies the meiotic chromosomes so they can be extracted without discarding the motor or centrosome molecules. Spindle collapse promises an efficient improvement, specifically, if oocyte recovery is complete and rapid.
Moreover, another embodiment of the present invention envisions using ooplasmic supplementation either by ooplast electrofusion (SCNT+OF) or microinjection (cytoplasmic transfer and ICNI; CT+ICNI), which has been used for bovine cloning and clinical ART (CT) (Barritt et al., 5 MOL. HUM. REPROD. 927-33 (1999)). Here additional ooplasm, from oocytes of the same clutch, supplements that lost during enucleation. An alternative embodiment may use the ooplasm from cold or nocodazole-recovery oocytes, for example, if the complete recovery within the same oocyte proves difficult. Ooplasmic supplementation has succeeded already in humans and cattle, and is particularly straightforward when combined with ICNI (i.e., ICNI+CT), just like the clinical ICSI+CT. ICS/NI-2PN (i.e., ICNI, ICSI and then pronuclear removal) uses half the oocytes of double NT. It demands a second day enucleation, but uses natural activation and avoids both cytochalasin and spindle disruption. The present invention has several important benefits for the biomedical research community. By expanding animal and specifically primate reproduction to include transgenic and SCNT capabilities, the utility of this model for essential and urgent preclinical investigations may be greatly enhanced. SCNT may find extraordinary applications, were it developed as a reliable, routine approach for propagating invaluable primate models. Notwithstanding the technologies routinely available for creating rodent models for various diseases, many serious human disorders are not appropriately studied in these lower mammals. The production of transgenic primates as the most clinically relevant models for human diseases might well be critical for the entire clinical research community. Furthermore, the combination of these approaches might even result in reliable and efficient applications for propagating invaluable transgenic primates as research models. Finally, the promise of safe and effective gene therapy protocols cannot be fully realized until an appropriate system for investigation is found to fill the gap between transgenic mice and seriously ill patients. Consequently, there are strong justifications for developing reliable and effective methods for creating genetically modified primates, specifically, human and non- human primates. Thus, the present invention may have clinical and investigative applications which include, but are not limited to, cell therapy (neural, brain, and spinal stem cell applications, liver stem cell applications, pancreas stem cell applications, cardiac stem cell applications, renal stem cell applications, blood stem cell applications, retinal stem cell applications, diabetes-stem cell applications, orthopedics-stem cell applications, identical primate models for research, drug discovery, embryonic stem cells for drug discovery), pharmaceutical and medical devices (including animal models of disease for drug discovery and testing, pharmacological target identification, drug discovery, drug efficacy testing, biocompatibility of medical devices), agriculture, rare and endangered species, and toxicology evaluation.
Furthermore, the present invention also relates to methods of using embryonic stem cells and transgenic embryonic stem cells to treat human diseases. Specifically, the methods for clonal propagation of primates, specifically, human or non-human primates, described in the present invention, may also be used to create embryonic stem cells and transgenic embryonic stem cells.
Cells from the inner cell mass of an embryo (i.e., blastocyst) may be used to derive an embryonic stem cell line, and these cells may be maintained in tissue culture (see, e.g., Schuldiner et al., 97 PROC. NATL. ACAD. SCI. USA 11307-12 (2000); Amit et al., 15 DEV. BlOL.271-78 (2000); U.S. Patent No. 5,843,780; and U.S. Patent No. 5,874,301). In general, stems cells are relatively undifferentiated, but may give rise to differentiated, functional cells. For example, hemopoietic stem cells may give rise to terminally differentiated blood cells such as erythrocytes and leukocytes.
Figure 3 provides the basic outline of such procedures, specifically, embryonic stem cells can grow into new nerves to heal injuries in a patient, such as spinal damage (Figure 3A). Eggs are removed from the patient's ovaries (Figure 3B) and placed in a petri dish. Cells that cling to the egg are removed (Figure 3C). Inside the egg is the nucleus (Figure 3D), which is removed in order to make a cloned embryo. For example, the egg is pierced with a fine needle or pipette (Figure 3E), gently squeezed or aspirated to expel the nucleus (Figure 3F), and the nucleus is removed (Figure 3G). One of the cells removed from the egg previously (Figure 3C) is injected into the egg (Figure 3H). An electric current activates the egg (Figure 31). The genetic material of the injected cell guides the egg to develop (Figure 3J). Cell division begins (Figure 3K). Specifically, the genetic material is dividing so that it can be shared equally among the two new cells. The cells have divided again, to the four-cell stage (Figure 3L). Many cell divisions later an area called the inner cell mass, which contains stem cells, is visible (Figure 3M). The stem cells are removed and placed in a growth medium (Figure 3N). The stem cells grow into colonies (Figure 30), which can be divided and grown repeatedly, resulting in millions of stem cells (Figure 3P). These cells can grow into any tissue in the primate body, for example, muscle, nerve, pancreas, and bone cells (Figure 3Q). Specialized cells may be placed back in a human patient at the site of injury or disease, so that new, working cells grow (Figure 3R).
Using the methods of the present invention, transgenic primate embryonic stem cells may also be produced which express a gene related to a particular disease. For example, transgenic primate embryonic cells may be engineered to express tyrosine hydroxylase, which is an enzyme involved in the biosynthetic pathway of dopamine. In Parkinson's disease, this neurotransmitter is depleted in the basal ganglia region of the brain. Thus, transgenic primate embryonic cells expressing tyrosine hydroxylase may be grafted into the region of the basal ganglia of a patient suffering from Parkinson's disease and potentially restore the neural levels of dopamine (see, e.g., Bankiewicz et al., 144 EXP. NEUROL. 147-56 (1997)). The methods described in the present invention, therefore, may be used to treat numerous human diseases and disorders (see, e.g., Rathjen et al., 10 REPROD. FERTIL. DEV. 31-47 (1998); Guan et al., 16 ALTEX 135-41 (1999); Rovira et al., 96 BLOOD 4111-117 (2000); Muller et al., 14 FASEB J. 2540-48 (2000)).
Other objectives, features, and advantages of the present invention will become apparent from the following specific examples. The examples, while indicating specific embodiments of the invention, are provided by way of illustration only. Accordingly, the present invention also includes those various changes and modifications within the spirit and scope of the invention that may become apparent to those skilled in the art from this detailed description.
EXAMPLES Example 1: Oocyte collection and Nuclear Transfer Procedures for superstimulation, oocyte staging, and fertilization of rhesus eggs have been described by Hewitson et al. (77 FERTIL. STERIL. 794-801 (2002)). Enucleation of mature unfertilized oocytes, exposed briefly to TALP-Hepes with 7.5 μg/ml cytochalasin D (CCD) and 1 μg/ml Hoechst 33342 DNA dye (Sigma Chemical Co., St. Louis, MO), was performed using a 30 μm ID pipette (Humagen, Charlottsville, VA) to aspirate the first polar body and underlying cytoplasm, including the second mitotic spindle (Meng et al., 57 BlOL. REPROD. 454-9 (1997)). Maternal chromatin removal was confirmed by Hoechst imaging. For embryonic cell nuclear transfer (ECNT), donor blastomeres were isolated from 4- to 32- cell embryos after zona pellucida removal with 0.5% pronase (Boehringer Mannheim) and culturing in Ca2+- and Mg^-free TALP-Hepes with 1 mM each EDTA and EGTA. A single blastomere was pipetted into the perivitelline space of an enucleated oocyte and, after a 30-60 min. recovery, fused with two DC pulses (1.2 kV/cm, 30 μsec) using a BTX Cell Manipulator 2001 (Genentronics, Inc., San Diego, CA) in 0.3 M sorbitol, 0.1 mM calcium-acetate, 0.1 mM magnesium acetate and 0.5 mg/ml FFA-BSA (Sigma). ECNTs were performed using activation prior to blastomere fusion 2-4 hours later, as well as aged or interphase oocytes for recipient cytoplasts. For SCNT, enucleated oocytes were either directly injected with a single donor cell or fused after transfer of a donor cell under the zona pellucida. The cell nuclear donor source included dissociated granulosa cells, endothelial cells collected from rhesus umbilical cords, isolated, cultured ICM cells derived from rhesus blastocysts (2-3 passages), and primary rhesus fibroblast cell lines.
Activation Protocols. NT constructs were activated between 1-4 hours after cell fusion to enable nuclear reprogramming either by microinjection of 120-240 pg ml-1 rhesus sperm extract prepared in 120 mM KCl, 20 mM HEPES, 100 μM EGTA, 10 mM sodium glycerophosphate (Wilmut et al., 419 Nature 583-87 (2002)) and sterilized through a 0.22 μm SpinX filter (Costar, Cambridge, MA) or by the sequential treatment of about 5 μM to about 10 μM ionomycin (5 min.; room temperature) and 1.9 mM DMAP for 4 hours (Chanet al., 287 SCIENCE 317-19 (2000)). After DMAP, eggs were washed extensively and cultured in TALP. FertClones were produced by the fusion of a cytoplast containing removed maternal chromatin with an enucleated oocyte and then fertilized by ICSI 2-3 hours later. All were cultured in TALP for 24 hours and then buffalo rat liver cell (BRL) monolayers in CMRL medium.
Microinjection. Antibody inhibition studies were performed using 9-μm micropipettes (Humagen) front-loaded with the primary antibody. Between 4-6% of the egg volume (~700 pi) was microinjected with antibodies at 2-10 mg ml-1. Final antibody concentrations were at between 70-350 pg total Ig protein per oocyte. For imaging microinjected oocytes and eggs, the primary antibody was omitted.
Embryo Transfer. For embryo transfer, surrogate rhesus females were selected on the basis of serum estradiol and progesterone levels. Pregnancies were ascertained by endocrinological profiles and fetal ultrasound performed between days 24-30 (Wilmut et al., 385 NATURE 810-3 (1997)). Imaging. Methods for fixation and immunocytochemistry of oocytes and NTs were performed as described. Mitalipov et al., 66 BIOL. REPROD. 1449-55 (2002). Centrosomes were detected with g-tubulin or pericentrin polyclonal rabbit antibodies (gifts of Dr. T. Stearns [Stanford] and S. Doxsey [U. of Mass], respectively). Thomson et al., 92 PROC. NATL. ACAD. SCI. USA 7844-48 (1995) and Thomson et al., 282 SCIENCE 1145-47 (1998). NuMA, HSET and Eg5 were detected using rabbit polyclonal antibodies as described. Wilmut et al. (1997), and Humpherys et al. (2002). Microtubules were immunolabeled with E7 mouse monoclonal antibody. Wilmut et al., 385 NATURE 810-3 (1997). Primary antibodies were detected using Alexa secondary antibodies (Alexa-546 goat anti-mouse IgG; Alexa-488 goat anti-rabbit IgG; Molecular Probes, Eugene, OR) for observation by confocal microscopy. Each primary and secondary antibody was applied for 40 min at 37°C; rinses performed with PBS + 0.1% Triton. DNA was fluorescently detected with 5μg/ml Hoechst 33342 (Sigma) and lμM Toto-3 (Molecular Probes) added to the penultimate rinse. Coverslips were mounted in Vectashield antifade (Vector Labs, CA). Controls included nonimmune and secondary antibodies alone, which did not detect spindle microtubules or centrosomes. Slides were examined using conventional immunofluorescence and laser- scanning confocal microscopy. Conventional fluorescence microscopy was performed first using a Nikon Eclipse ElOOO microscope with high numerical aperture objectives. Data was recorded digitally using a cooled CCD camera (Hamamatsu Instruments Inc., Japan) using Metamorph software (Universal Imaging, West Chester, PA). Laser-scanning confocal microscopy was performed using a Leica SP-2 equipped with Argon and Helium-Neon lasers for the simultaneous excitation of Alexa-conjugated secondary antibodies and Toto-3 DNA stain.
Immunoblotting. Murine oocytes and human ovarian protein extracts (Clontech, Inc., Palo Alto, CA) were separated on linear gradient SDS-PAGE gels and Western blots as described by Humpherys et al. (2002).
Example 2. Production of Embryonic Stem Cells
ES cells are established from embryos by the following method. Following SCNT, 2- 4 blastomeres are cultured in a microwell, which contains a monolayer of feeder cells derived from mouse embryonic fibroblasts (MEF), or primate embryonic fibroblasts (PEF), either human or non-human (Richards et al., 21(5) Stem Cells 546-56 (2003); Richards et al., 20
Nature Biotech. 933-36 (2002)). The remaining embryo is then transferred to an empty zona for embryo reconstruction as described in Example 1. This co-culture system for isolating and culturing an ES cell line is well known in the art (see, e.g., Thomson et al., 92 Proc. Natl. Acad. Sci. USA 7844-48 (1995); Ouhibi et al., 40 MoI. Reprod. Dev. 311-24 (1995)). It has been suggested that the feeder cells provide growth factor-like leukemia inhibiting factor (LIF), which inhibits stem cell differentiation. The microwells contain 5-10 μl of culture medium (80% DMEM as a basal medium, 20% FBS, ImM β-mercaptoethanol, 1000 units/ml LIF, non-essential amino acids, and glutamine). The cells are then incubated at 370C with 5 % CO2 and covered with mineral oil. Fresh medium is replaced everyday and the survival of blastomeres is determined by cell division. During the initial culture, cell clumps are dissociated mechanically until cell attachment to the MEF monolayer and colony formation is observed. The colonies are then passaged to a 4-well plate and subsequently to a 35 mm dish in order to expand the culture gradually until a stable cell line is established. In addition to the dissociated blastomere culture, the reconstructed embryos are also cultured until the blastocyst stage is reached. Hatch blastocysts or blastocysts without zonae are cultured on a MEF monolayer in a microwell as described above. Instead of dissociating the blastomeres, the blastocysts are allowed to attach to the MEF monolayer. Once the blastocysts attach to the MEF, the ICM cells are isolated mechanically and transferred to a fresh culture well. The embryonic cells are cultured as described above and expansion of the cells is continued until individual colonies are observed. Individual colonies are selected for clonal expansion. This clonal selection and expansion process continues until a clonal cell line is established.
Infection of unfertilized oocytes by a pseudotyped retroviral vector has been used successfully to produce a transgenic non-human primate. These methods are disclosed in co- pending U.S. Patent Applications Serial No. 09/736,271 and Serial No. 09/754,276, which are expressly incorporated herein by reference. The presence of the transgene was demonstrated in all tissues of the transgenic monkey, which suggests an early integration event has occurred, perhaps in the maternal chromosome prior to fertilization. To produce a transgenic embryonic stem cell line, the transgenic embryos produced by pseudotype infection are dissociated as described above in the clonal embryo production process. These split embryos are then used to produce clonal offspring or its embryonic counterpart is used to produce a transgenic embryonic stem cell line. Thus, the transgenic offspring and the transgenic embryonic stem cell line share the same genetic modification that was achieved at the oocyte stage. Various modifications and variations of the described examples and systems of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention, which are obvious to those skilled in related fields, are intended to be within the scope of the following embodiments. Example 3. Oocyte collection. Procedures for superstimulation, oocyte staging, and intracytoplasmic sperm injection (ICSI) fertilization of nonhuman primate eggs have been described by Hewitson, 13 HUM. REPROD. 3449-3455 (1998). Immature bovine oocytes were obtained from Bomed (Madison, WI) and overnight express shipped in modified TC 199 maturation media for the collection of mature oocytes as described previously. Navara, 162 DEV. BIOL. 29-40 (1994). Enucleation. Removal of the meiotic spindle and chromosomes was accomplished in two ways. Using the 'squish' enucleation method (Hwang, 303 SCIENCE 1669-1674 (2004)) for pre-Metaphase II spindle aspiration, a cumulus-free oocyte was held with a holding micropipette (110-μm inner diameter) and the zona pellucida was partially dissected with a fine glass needle to make a slit near the first polar body. The first polar body and adjacent cytoplasm containing the meiotic spindle, ranging from telophase-I to prometaphase-II, were extruded by squeezing with the needle. Oocytes were enucleated in Hepes-buffered TALP supplemented with 0.3% BSA and 7.5 μg/ml cytochalasin B (Sigma-Aldrich Corp, St. Louis, MO). Alternatively, for Metaphase-II spindle aspiration by extraction, mature unfertilized oocytes were first exposed to TALP-Hepes with 7.5 μg/ml cytochalasin D (CCD; Sigma Chemical Co., St. Louis, MO) prior to using suction applied by a 22 μm LD pipette (Humagen, Charlottesville, VA) to aspirate the first polar body and underlying cytoplasm, including the second meiotic spindle. After enucleation, karyoplasts were stained with 5 μg/ml bisbenzimide (Hoechst 33342, Sigma-Aldrich Corp.) for 5 min and observed under an inverted microscope equipped with epifluorescence. Oocytes still containing DNA materials were excluded from further experiments.
Nuclear Transfer. The somatic cell nuclear donor source included dissociated cumulus cells (both autologous and heterologous sources) and primary rhesus fibroblast cell lines. Trypsinized single cells from the confluent fibroblast line demonstrating a smooth surface or cumulus cells were selected under an inverted microscope and transferred into the perivitelline space of enucleated oocytes. These couplets were equilibrated with 0.3 M mannitol solution containing 0.5 mM Hepes, 0.1 mM CaC12, and 0.1 mM MgC12 for 4 minutes and transferred to a chamber containing two electrodes that were overlaid with the mannitol solution. Couplets were fused with two DC pulses of 2.7 kV/cm for 15 μsec using a BTX Electro-Cell Manipulator 2001 (BTX, Inc., San Diego, CA). Successful fusion was confirmed 45-60 minutes after electroporation by absence of the donor cell in the perivitelline space.
For ECNT (embryonic blastomere cell nuclear transfer), donor blastomeres were isolated from 16-to-32-cell stage embryos after zona pellucida removal with 0.5% pronase (Boehringer Mannheim, Indianapolis IN) and culturing in Ca++- and Mg++-free TALP- Hepes with 1 mM each EDTA and EGTA. A single blastomere was pipetted into the perivitelline space of an enucleated oocyte and, after a 30-60 min recovery, fused with two DC pulses as described above. For some cumulus SCNT' s, enucleated oocytes were directly injected with a single donor cell using a 9-μm pipet, following the mouse SCNT procedures of intracytoplasmic nuclear injection (ICNI). Simerly & Navara, 5 CLONING STEM CELLS 319-331 (2003); Wakayama & Perry, PRINCIPLES OF CLONING 301-341 (2002). Bovine SCNT and ECNT was performed as previously described Navara, 162 DEV. BlOL. 29^10 (1994). Activation Protocols. Reconstructed embryos were activated (Table I) by: i. simultaneously electrical fusion treatment (EFA); ii. chemical activation 2 hours after NT fusion by exposure to 5 μM ionomycin (CalBiochem, La Jolla, CA) for 4 minutes followed by a 4 hour incubation in 1.9 mM 6-dimethylaminopurine (EFIAD) or iii. injection of NHP sperm extract [EFSFA; 120-240 pg ml-1]. Simerly & Navara, supra. Activated oocytes were washed three times with TALP-HEPES (Bavister, 28 BIOL. REPROD. 983-999 (1983)) supplemented with 4 mg/ml fatty acid-free BSA (Sigma-Aldrich Corp.) and placed in 25 μl microdrops (5-7 oocytes per drop) of G 1.1 culture media (Vitrolife, Boulder, CO) under mineral oil at 37°C in 5% CO2.
Fertilization Achieved by Mimicking Cloning Procedures. To dissect the adverse consequences of SCNT' s many harsh physical and chemical procedures (e.g. cytochalasin, physical enucleation, electrical fusion, chemical activation, etc.) from the biological incompatibilities of SCNT itself, fertilized eggs were reconstructed using the procedures of cloning. These FertClones were produced by the fusion of a donor nucleus into an intact oocyte followed by ICSI fertilization 2-3 hours later. SCNT + met-II intact constructs were produced by fusion of the donor nucleus into an intact oocyte followed by artificial activation with either sperm factor or ionomycin/DMAP as described above.
In vitro culturing. All nuclear transfer constructs were developed in sequential Gl- G2-mSOF media, as described by Hwang et al., supra, which is hereby incorporated by reference. Gl media was employed for 48 hours after NT prior to transfer to G2 media for an additional 48 hours. Around the morula stage, embryos were transferred to mSOF with fructose until blastocyst stage.
Embryo Transfer. For embryo transfer, surrogate macaque females were selected on the basis of serum estradiol and progesterone levels. Pregnancies were ascertained by endocrinological profiles and fetal ultrasound performed between days 16-30 Hewitson, 5 NAT. MED. 431 433 (1999).
Imaging. Live oocyte imaging was accomplished using a Nikon TE 2000 inverted microscope equipped with polarization optics (SpindleView™; CRI, Cambridge, MA) and a thermoplate temperature control stage (Tokai HIT Inc, Japan) to maintain 37°C. Methods for immunocytochemistry were performed as described in Simerly & Schatten, 225 METHODS ENZYMOL. 516-553 (1993). NuMA, HSET (human spleen embryonic tissue and testis), and Eg5 were detected using rabbit polyclonal antibodies as described in Mountain, 147 J. CELL BIOL. 351-366 (1999); Simerly et al., 300 SCIENCE, 297 (2003). Microtubules were immunolabeled with E7 mouse monoclonal antibody. Simerly, 1 NAT. MED. 47-52 (1995). BrDU labeling was performed using a kit according to the manufacturer's instructions as previously published. Hewitson, supra. Primary antibodies were detected using Alexa secondary antibodies (Alexa-546 goat anti-mouse IgG; Alexa-488 goat anti-rabbit IgG; Molecular Probes, Eugene, OR). Each primary and secondary antibody was applied for 40 minutes at 37°C. Rinses were performed with PBS + 0.1% Triton. DNA was fluorescently detected with lOμg/ml Hoechst 33342 (Sigma) and lμM Toto-3 (Molecular Probes) added to the penultimate rinse. Coverslips were mounted in Vectashield antifade (Vector Labs, Burlingame, CA). Controls included nonimmune and secondary antibodies alone, which did not detect spindle microtubules or centrosomes. Slides were examined using conventional immunofluorescence and/or laser-scanning confocal microscopy. Conventional fluorescence microscopy was performed first using a Nikon Eclipse ElOOO microscope with high numerical aperture objectives. Data was recorded digitally using a cooled CCD camera (Hamamatsu Instruments Inc., Japan) using Metamorph software (Universal Imaging, West Chester, PA). Laser-scanning confocal microscopy was performed using a Leica SP-2 equipped with Argon and Helium-Neon lasers for the simultaneous excitation of Alexa- conjugated secondary antibodies and Toto-3 DNA stain.
Results 'Squish' enucleation and in vitro development of SCNT NHP constructs.
Dynamic imaging of the spindle-chromosome complex (SCC) in live unfertilized NHP oocytes by SpindleView™ polarization optics is useful for the accurate assessment of meiosis prior to enucleation. The second meiotic spindle is seen as a bright, birefringent bipolar structure against the dark cytoplasm (Fig. 4A, arrow). 'Squish' enucleation of oocytes that nearly complete first polar body extrusion, often removed the SCC at the telophase-I stage (Fig. 4B). Following enucleation by extrusion at this pre-metaphase-II stage, a single rhesus fibroblast cell demonstrating a normal karyotype (Fig. 4C, inset) was transferred by electrofusion, which also served to simultaneously activate the constructs. By 8 hours post- fusion and activation, a single decondensed nucleus was visible in the smooth cytoplasm of the 1-cell construct (Fig. 4C).
In vitro preimplantation development from first cleavage to expanded, hatched blastocyst stage is shown in Fig. 4D-K and Table I. The in vitro development of SCNT- derived constructs is accelerated compared to ICSI fertilized embryo development. Hewitson, supra. By 24 hours post activation, the SCNT constructs have undergone first cleavage, showing even blastomeres with a single nucleus apparent in each daughter cell (Fig. 4D). By 36 hours post-activation, second division commences (Fig 4E; 3-cell embryo with a single daughter nucleus visible in upper left blastomere). At 50 hours post-activation, 8-cell SCNT embryos were observed, with even sized, smooth blastomeres and single nuclei within each cell (Fig. 4F). Slight fragmentation, a common occurrence in NHP embryos, is also visible in the perivitelline space of some embryos (Fig. 4F, arrow). Development to the 16-cell stage was observed by 72 hours post-activation (Fig. 4G; focused on upper plan to show single nuclei in each blastomere) and by 96 hours post activation, the compacted morula with flattened, undistinquished blastomeres was observed (Fig.4H). The first signs of the early blastocyst (Fig. 41) containing a conspicuous blastocoel (arrow) is found by 110 hours after activation and the expanded blastocyst with a large, distended blastocoel cavity observed by 125 hours post-activation (Fig. 4J). The fully expanded, hatched blastocyst was observed by 154 hours post activation, the stage typically selected for isolation of the ICM to derived stem cells (Fig. 4K). Table I presents NHP SCNT development with either fresh, homologous cumulus cells or confluent heterologous rhesus skin fibroblast cells. Higher successful NT fusion, nuclear reformation and embryonic development were observed using the fibroblast cells as a donor source as compared to cumulus cells. Furthermore, when the electrofusion intensity permits simultaneous fibroblast fusion and cytoplast activation (EFA), significantly more expanded blastocysts were produced compared to either chemical or sperm factor activation after 2 hours of incubation post-NT. Of the three SCNT blastocyst produced by simultaneous fusion/activation, two had identifiable inner cell mass [ICM] cells following immunosurgery for NT-nhp-ES cell derivations. A fourth chimeric blastocyst was derived by the reaggregation of an SCNT morula (electrofused followed by 2 hour ionomycin/DMAP isolation) with a fertilized morula (Fig. 4L). AU of these putative NT-nhp-ES cells stopped growing in vitro after one week.
No evidence of a sustained pregnancy was confirmed (Table II). Early pregnancy indicators, e.g. ultrasonography and serum nhpCG measurements around day 16, are known to be inconclusive predictors for on-going pregnancies. Munro, 41 AM. J. PRIMATOL. 307- 322 (1997). Table II reports pregnancy establishments as verified by fetal heartbeats imaged at day 30.
Few SCNT embryos (-1%) develop to the blastocyst stage. See Mitalipov, 66 BlOL. REPROD. 1367-1373 (2002). Here, despite SCNT embryos appearing morphologically normal (Fig. 4C-K; Table I), many NT constructs generated by aspirated enucleation still demonstrate aneuploidy (Fig. 5). DNA mis-segregation is prevalent by first mitotic telophase (Fig. 5A), as evident by lagging chromosomes and loosely organized DNA at the spindle poles (Fig. 5A, blue, DNA; arrow: lagging chromatin). At second division (Fig. 5B), numerous karyomeres in daughter blastomeres (blue) and abnormal assembly of the second mitotic spindles was observed (green). By the 4-cell stage (Fig. 5C), the DNA configurations (blue) vary widely within each daughter blastomere. As development proceeded to the 8-cell stage (Fig. 5D-5E), monastral interphase microtubules patterns (green) either lacking DNA or with inappropriate chromosomes (blue) was observed. A control parthenogenetic embryo at the 8-cell stage demonstrates typical microtubule and DNA configurations for this stage (Fig. 5F).
Abnormal first interphase microtubule organization in NHP SCNT or ECNT constructs was distinct from bovine NT clones or NHP androgenotes. The somatic cell's nuclear DNA and cytoplasmic constituents [i.e. mitochondria and the centrosome, the cell's major microtubule organizing center (MTOC)], entered the oocyte's cytoplasm after SCNT by fusion. Analysis of microtubule patterns in SCNT constructs, made by aspiration of mature metaphase-II meiotic spindles, showed that the somatic centrosome ineffectively assembles cytoplasmic microtubules near the incorporated somatic nucleus by the end of interphase (Fig. 6A and 6B: microtubules, green; DNA, blue), if at all. Typically, a cortical disarrayed microtubule pattern was observed after SCNT construct activation (Fig. 6C: microtubules, green; DNA, blue; Wu, 55 BIOL. REPROD. 260-270 (1996)). After ECNT, the contributed blastomere centrosome(s) consistently assembled cytoplasmic microtubules (ECNT; Fig. 6D, arrows). Conversely, when NT was performed using a male pronucleus from an ICSI fertilized zygote as the donor, a symmetrical microtubule array emanating from the sperm centrosome was observed (Fig. 6E, MPn, arrow). Following SCNT into a metaphase-II intact oocyte and activation by ICSI fertilization [Fert+SCNT], BrDU nuclear labeling indicated successful DNA synthesis onset and cell cycle progression in the transferred somatic nucleus, as observed in the decondensed male and female pronuclei, respectively (Fig. 6F, BrDU, green).
Interspecific SCNT. Because bovine SCNT succeeded at significantly higher rates than NFIP SCNT, interspecific SCNT was performed to address the issue as to whether the bovine cytoplasm was more conducive to NT-spindle formation than the NHP ooplasm, or whether the bovine somatic cell's centrosome was more resilient than the NHP somatic centrosome for NT-spindle organization. Unlike non-human primate clones, bovine X bovine SCNT results in the formation of a single radially arrayed aster juxtaposed to the nucleus, akin to fertilization's sperm aster (SCNT: Fig 6G; ECNT: Fig. 6H; microtubules, green). Bovine NT constructs produced normal bipolar spindles with aligned chromosomes at first mitosis (Fig. 61). Furthermore, the bovine egg cytoplasm appeared to tolerate exogenous centrosome transfer better than NHP constructs, as evident by a focused microtubule astral array adjacent to the reformed fibroblast nucleus after SCNT into the bovine enucleated cytoplast (Fig. 6J).
Androgenesis. We examined microtubule and DNA patterns in androgenotes, derived by ICSI fertilization of enucleated oocytes (Fig. 7). As the introduced sperm DNA decondensed in the enucleated cytoplast, a microtubule astral array assembled from the introduced paternal centrosome (Fig 7A, arrow; arrowhead: incorporated sperm axoneme). The microtubules expanded within the cytoplasm (Fig. 7B, green) and, at late interphase, were observed emanating from the duplicated, split paternally-derived centrosomes adjacent to the fully formed male pronucleus (Fig. 7C, arrows). At late prometaphase, a bipolar spindle with asters (centrosomes) at each pole and aligning metaphase chromosomes assembled (Fig. 7D, arrows; arrowhead: sperm axoneme). Androgenotes typically progressed through first mitosis, completing accurate chromosome segregation at the first cell division (Fig. 7E). 80% [8/10] of androgenotes examined at the pronuclear stage demonstrated normal microtubule and chromosome patterns, though only 24% [8/33] produced normal 8-cell stage embryos.
SCNT clones, missing spindle molecular motors and matrix proteins, form multipolar spindles with misaligned chromosomes. At first mitosis, NT constructs, generated after aspiration of the mature metaphase II meiotic spindle, developed disarrayed spindles with misaligned chromosomes (Fig 8A-8C; see also Simerly et al., supra. Following blastomere NT, the MTOCs observed nucleating microtubules were distinctly misaligned on the assembled tri- or tetrapolar metaphase spindles containing scattered chromosomes (Fig 8A and 8B, green, arrows). Similar aberrant mitotic spindle assembly and chromosomal anomalies were found in mitotic SCNT' s, though definitive spindle pole MTOCs that nucleate microtubules were generally lacking (Fig. 8C, green).
Key centrosomal and molecular motor proteins were deficient in mitotic spindles after nuclear transfer, compared with spindle assembly in constructs produced by combining nuclear transfer with either fertilization (Simerly et al., supra) or artificial activation of intact oocytes (i.e., no enucleation of the SCC). NuMA, a spindle pole assembly protein found in mammalian oocytes (Lee, 62 BIOL. REPROD. 1184-1192 (2000); Tang, 11 J. BIOMED. SCI. 370-376 (2004)) concentrated in the reconstituted interphase SCNT or ECNT nucleus following activation (Fig. 8D, green). However, by first mitosis, NuMA protein was either not detected (Simerly et al., supra) or at barely detectable concentrations on the NT mitotic spindle poles (Fig. 8E; green). Conversely, mitotic constructs produced from the activation of intact oocytes after SCNT (SCNT + Met-II) generated spindles with more aligned chromosomes and abundant NuMA on the poles, but still with misaligned MTOCs (Fig. 8F, green; arrows: centrosomes). Similarly, the mitotic kinesin HSET, which traffics to the microtubule minus-ends in meiotic and mitotic spindles (Simerly et al., supra) was undetected in mitotic nuclear transfer spindles displaying misaligned chromosomes (Fig. 8G; green; inset: microtubules, red; DNA, blue). HSET protein detection was restored at spindle poles of mitotic SCNT + Met-II constructs (Fig. 8H, green), though the chromosomes remained scattered in the microtubule spindle lattice (Fig. 8H, blue). The oppositely directed kinesin mitotic microtubule motor protein Eg5, observed at the centromeres and on the kinetochore-microtubule bundles in meiotic and mitotic spindles (Simerly et al., supra), remained paired at sister kinetochores on multipolar spindles with misaligned chromosomes in mitotic nuclear transfers (Fig. 81). HSET and NuMA detection in the enucleated karyoplast and in the remaining cytoplast following removal of the SCC was investigated. Following successful enucleation of the SCC (Fig 9A, inset, DNA), no assembled microtubules were observed in the cytoplast (Fig. 9A, green). A large monastral interphase microtubule array assembled following sperm factor or ionomycin/DMAP chemical activation of the enucleated cytoplast (Fig. 9B, green; inset: DNA). Rarely, activated cytoplasts will produce atypical, astral bipolar spindle-like structures without DNA at first mitosis (Fig. 9C, green; inset: DNA). The SCC- containing karyoplast, produced by 'squish' enucleation, retained the intact metaphase-II spindle (Fig. 9D; inset: red, microtubules; blue, DNA) with HSET protein detected in the spindle lattice (Fig. 9D, green). Cumulus and fibroblast SCNT donor cells retained somatic HSET at the cell cycle stage used for NT (Fig. 9E and 9F, green).
Taxol-induced microtubule recruitment. Paclitaxel, an effective microtubule disassembly inhibitor, was employed to investigate cytoplasmic HSET recruitment to augmented microtubules in the presence or absence of the SCC. Exposure of intact oocytes to Paclitaxel enhanced HSET protein only at the second meiotic spindle poles (Fig 9G: arrowheads, HSET, green: arrows: cytoplasmic microtubule asters). When the enucleated cytoplast lacking the SCC (Fig 9H, blue, DNA) was treated with paclitaxel, numerous cytoplasmic microtubule bundles assembled in the cytoplasm (Fig. 91, red), but without HSET protein detection (Fig. 9 J, green).
Cytoplasmic NuMA after removal of the SCC. NuMA resided at the spindle poles of the SCC following enucleation by extrusion (Fig. 1OA, green). The remaining enucleated cytoplast had neither assembled microtubules (Fig. 1OB, upper inset, red) nor detectable cytoplasmic NuMA (Fig. 1OB, green; lower inset: DNA). Paclitaxel treatment of SCC- intact oocytes showed NuMA localization at disorganized spindle poles (Fig. 1OC, green, lower arrow) and the assembled cytoplasmic microtubule asters (Fig. 1OC, green, arrowhead). Both cumulus and fibroblast SCNT donor cells demonstrated intranuclear NuMA prior to NT (Fig. 1OD and 1OE, green). Yet, abundant maternal NuMA protein remained after SCC extrusion and was available for nuclear importation,. Furthermore, the amount of somatic intranuclear NuMA provided by a fibroblast cell at fusion was relatively small compared to the maternal protein available for nuclear importation (Fig. 1OF; green, arrowhead: nonfused fibroblast cells). Nevertheless, NuMA recruitment to NT spindles following SCNT appeared compromised. Failed-to-activate SCNT + fertilization constructs which remain firmly arrested at second meiosis often display NuMA at both meiotic spindle poles (Fig. 1OG; arrowheads) and at the condensed sperm head (Fig. 1OG; arrow), but with NuMA missing from the chaotic spindle surrounding the transferred nucleus (Fig. 1OG, NT). Conversely, SCNT combined with aging metaphase-II intact oocytes (Fig. 1OH: upper arrow) that subsequently failed to activate, assembled bipolar NT spindles that recruited cytoplasmic NuMA to their poles, though chromosomes do not align properly (Fig. 1OH, lower arrow: green, NuMA).
Table I: Development of NHP clones reconstructed with fresh cumulus cells or skin fibroblasts.
Number of cloned embryos to develop (% of fused)
No. (%)of oocytes
Donor cells Fusion No. 2-4 NT+Fert (source) activation Enucleated Fused PN formation cell 8-16 cell chimerae Morula Blastocyst
EF
15 12 (80) 6 (50) 3 (50) 2 (33) 2 (33) 1 (17)
Cumulus Aa
(homologous)
EFIADb 14 10 (71) 6 (60) 5 (83) 4 (67) 0 0
Figure imgf000038_0001
dOne morula was the NT+Fert chimera, with one resulting blastocyst
U)
Table II: Outcomes After SCNT Transfers into Timed Recipients.
Figure imgf000038_0002

Claims

What is claimed is:
1. A method comprising the steps of: introducing nuclei along with one or more molecular components into an extrusion- enucleated egg, thus creating a nuclear transfer construct; culturing said nuclear transfer construct to produce a viable embryo; transferring said embryo to the oviducts of a female; and producing a cloned animal.
2. The method of claim 1, wherein said enucleated egg is a cumulus-free oocyte.
3. The method of claim 1, wherein said enucleated egg is enucleated pre-metaphase II.
4. The method of claim 1, wherein said enucleated egg is enucleated just prior to metaphase II arrest.
5. The method of claim 1, wherein said extrusion comprises: holding said egg with a holding micropipette; partially dissecting the zonal pellucida of said egg with a needle by making a slit near the first polar body of said egg; extruding the first polar body and adjacent cytoplasm containing the meiotic spindle, ranging from about telophase-I to about pro-metaphase-II, by squeezing said needle.
6. The method of claim 5, wherein said holding micropipette has an about 110 μm inner diameter.
7. The method of claim 5, wherein said needle is a glass needle.
8. The method of claim 1, wherein said egg is enucleated in Hepes-buffered TALP supplemented with BSA and cytochalasin B.
9. The method of claim 1, wherein said egg is enucleated in Hepes-buffered TALP supplemented with about 0.3% BSA and about 7.5 μg/ml cytochalasin B.
10. The method of claim 1, wherein said nuclei are derived from a somatic cell nuclear donor source.
11. The method of claim 10, wherein said nuclei derived from said somatic cell nuclear donor source include dissociated cumulus cells.
12. The method of claim 11, wherein said dissociated cumulus cells are autologous.
13. The method of claim 11, wherein said dissociated cumulus cells are heterologous.
14. The method of claim 11, wherein said dissociated cumulus cells are autologous and heterologous.
15. The method of claim 1, wherein said nuclei are derived from primary rhesus fibroblast cell lines.
16. The method of claim 1, wherein said nuclei are derived from donor blastomeres.
17. The method of claim 1, wherein said nuclei are transferred into the perivitelline space of said enucleated egg.
18. The method of claim 1, wherein said nuclear transfer constructs are equilibrated with mannitol solution.
19. The method of claim 18, wherein said mannitol solution comprises about 0.3 M mannitol solution containing about 0.5 mM Hepes, about 0.1 mM CaCl2, and about 0.1 mM MgCl2.
20. The method of claim 18, wherein said nuclear transfer constructs are equilibrated with said mannitol solution for about 4 minutes.
21. The method of claim 18, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing an electrode overlaid with said mannitol solution.
22. The method of claim 18, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing electrodes overlaid with said mannitol solution.
23. The method of claim 22, wherein said chamber contains two electrodes overlaid with said mannitol solution.
24. The method of claim 1, wherein said nuclei and said egg are fused with two DC pulses.
25. The method of claim 24, wherein said DC pulses constitute about 2.7 kK/cm.
26. The method of claim 24, wherein the duration of said DC pulses is about 15 μs.
27. The method of claim 1, wherein said nuclear transfer construct is developed in culture media.
28. The method of claim 27, wherein said culture media includes Gl, G2, and modified synthetic oviductal fluid (mSOF).
29. The method of claim 27, wherein said nuclear transfer construct is developed in said culture media sequentially.
30. The method of claim 27, wherein said nuclear transfer construct is developed in Gl for about 48 hours after nuclear transfer.
31. The method of claim 27, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours.
32. The method of claim 27, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours and transferred to mSOF around the morula stage until said nuclear transfer construct reaches the blastocyst stage.
33. The method of claim 27, wherein said mSOF media further comprises fructose.
34. The method of claim 1, wherein said nuclei have desired characteristics.
35. The method of claim 34, wherein said desired characteristics are linked to a specific disease or disorder.
36. The method of claim 35, wherein said specific disease or disorder is selected from the group consisting of cardiovascular disease, neurological disease, reproductive disorder, cancer, eye disease, endocrine disorder, pulmonary disease, metabolic disorder, autoimmune disorder, and aging.
37. The method of claim 1, wherein said introducing step comprises performing SCNT.
38. The method of claim 37, further comprising the step of performing pronuclear removal after SCNT.
39. The method of claim 37, further comprising the step of performing a second nuclear transfer following said SCNT.
40. The method of claim 1, wherein said introducing step further comprises performing meiotic spindle collapse.
41. The method of claim 1, further comprising the step of performing ooplasmic supplementation following said introducing step.
42. The method of claim 41, wherein said ooplamsic supplementation is performed by ooplast electrofusion.
43. The method of claim 41, wherein said ooplasniic supplementation is performed by microinjection.
44. The method of claim 1, wherein said one or more molecular components comprise centrosomal components normally present in sperm centrosomes.
45. The method of claim 1, wherein said one or more molecular components comprise mitotic motor proteins and centrosome proteins.
46. The method of claim 45, wherein said mitotic motor proteins comprise kinesins.
47. The method of claim 46, wherein said kinesins comprise HSET kinesin.
48. The method of claim 45, wherein said centrosome proteins comprise NuMA.
49. The method of claim 1, wherein said animal is a primate.
50. The method of claim 49, wherein said animal is a non-human primate.
51. The method of claim 50, wherein said non-human primate is a monkey.
52. The method of claim 49, wherein said primate is a human.
53. The method of claim 1, wherein said viable embryo is transgenic.
54. The method of claim 1, further comprising the step of producing embryonic stem cells from said viable embryo.
55. The method of claim 54, wherein said embryonic stem cells are human and said viable embryo is human.
56. An animal produced by the method of claim 1.
57. The animal of claim 56, wherein said animal is a primate.
58. The animal of claim 57, wherein said primate is a non-human primate.
59. The animal of claim 57, wherein said primate is a human.
60. A method comprising the steps of: introducing nuclei along with one or more molecular components into an extrusion- enucleated egg, thus forming a nuclear transfer construct; culturing said nuclear transfer construct to produce a viable embryo; dissociating blastomeres from said embryo; and culturing said blastomeres to produce stem cells.
61. The method of claim 60, wherein said enucleated egg is a cumulus-free oocyte.
62. The method of claim 60, wherein said enucleated egg is enucleated pre-metaphase II.
63. The method of claim 60, wherein said enucleated egg is enucleated just prior to metaphase II arrest.
64. The method of claim 60, wherein said extrusion comprises: holding said egg with a holding micropipette; partially dissecting the zonal pellucida of said egg with a needle by making a slit near the first polar body of said egg; extruding the first polar body and adjacent cytoplasm containing the meiotic spindle, ranging from about telophase-I to about pro-metaphase-II, by squeezing said needle.
65. The method of claim 64, wherein said holding micropipette has an about 110 μm innter diameter.
66. The method of claim 64, wherein said needle is a glass needle.
67. The method of claim 60, wherein said egg is enucleated in Hepes-buffered TALP supplemented with BSA and cytochalasin B.
68. The method of claim 60, wherein said egg is enucleated in Hepes-buffered TALP supplemented with about 0.3% BSA and about 7.5 μg/ml cytochalasin B.
69. The method of claim 60, wherein said nuclei are derived from a somatic cell nuclear donor source.
70. The method of claim 69, wherein said nuclei derived from said somatic cell nuclear donor source include dissociated cumulus cells.
71. The method of claim 70, wherein said dissociated cumulus cells are autologous.
72. The method of claim 70, wherein said dissociated cumulus cells are heterologous.
73. The method of claim 70, wherein said dissociated cumulus cells are autologous and heterologous.
74. The method of claim 60, wherein said nuclei are derived from primary rhesus fibroblast cell lines.
75. The method of claim 60, wherein said nuclei are derived from donor blastomeres.
76. The method of claim 60, wherein said nuclei are transferred into the perivitelline space of said enucleated egg.
77. The method of claim 60, wherein said nuclear transfer constructs are equilibrated with mannitol solution.
78. The method of claim 77, wherein said mannitol solution comprises about 0.3 M mannitol solution containing about 0.5 mM Hepes, about 0.1 mM CaCl2, and about 0.1 mM MgCl2.
79. The method of claim 77, wherein said nuclear transfer constructs are equilibrated with said mannitol solution for about 4 minutes.
80. The method of claim 77, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing an electrode overlaid with said mannitol solution.
81. The method of claim 77, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing electrodes overlaid with said mannitol solution.
82. The method of claim 81, wherein said chamber contains two electrodes overlaid with said mannitol solution.
83. The method of claim 60, wherein said nuclei and said egg are fused with two DC pulses.
84. The method of claim 83, wherein said DC pulses constitute about 2.7 kK/cm.
85. The method of claim 83, wherein the duration of said DC pulses is about 15 μs.
86. The method of claim 60, wherein said nuclear transfer construct is developed in culture media.
87. The method of claim 86, wherein said culture media includes Gl, G2, and modified synthetic oviductal fluid (mSOF).
88. The method of claim 86, wherein said nuclear transfer construct is developed in said culture media sequentially.
89. The method of claim 86, wherein said nuclear transfer construct is developed in Gl for about 48 hours after nuclear transfer.
90. The method of claim 86, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours.
91. The method of claim 86, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours and transferred to mSOF around the morula stage until said nuclear transfer construct reaches the blastocyst stage.
92. The method of claim 86, wherein said mSOF media further comprises fructose.
93. The method of claim 60, wherein said nuclei have desired characteristics.
94. The method of claim 93, wherein said desired characteristics are linked to a specific disease or disorder.
95. The method of claim 94, wherein said specific disease or disorder is selected from the group consisting of cardiovascular disease, neurological disease, reproductive disorder, cancer, eye disease, endocrine disorder, pulmonary disease, metabolic disorder, autoimmune disorder, and aging.
96. The method of claim 60, wherein said introducing step comprises performing SCNT.
97. The method of claim 96, further comprising the step of performing pronuclear removal after said SCNT.
98. The method of claim 96, further comprising the step of performing a second nuclear transfer following said SCNT.
99. The method of claim 60, wherein said introducing step further comprises performing meiotic spindle collapse.
100. The method of claim 60, further comprising the step of performing ooplasmic supplementation following said introducing step.
101. The method of claim 100, wherein said ooplamsic supplementation is performed by ooplast electrofusion.
102. The method of claim 100, wherein said ooplasmic supplementation is performed by microinjection.
103. The method of claim 60, wherein said one or more molecular components comprise centrosomal components normally present in sperm centrosomes.
104. The method of claim 60, wherein said one or more molecular components comprise mitotic motor proteins and centrosome proteins.
105. The method of claim 104, wherein said mitotic motor proteins comprise kinesins.
106. The method of claim 105, wherein said kinesins comprise HSET kinesin.
107. The method of claim 104, wherein said centrosome proteins comprise NuMA.
108. The method of claim 60, wherein said stem cells are primate embryonic stem cells.
109. The method of claim 108, wherein said embryonic stem cells are transgenic primate embryonic stem cells.
110. The method of claim 109, wherein said transgenic embryonic stem cells are transgenic non-human primate embryonic stem cells.
111. An embryonic stem cell produced by the method of claim 60.
112. The embryonic stem cell of claim 111, wherein said embryonic stem cell is used for gene therapy.
113. The embryonic stem cell of claim 111, wherein said embryonic stem cell is used as a therapy for human disease.
114. A method comprising the steps of: introducing nuclei along with one or more molecular components into an extrusion- enucleated egg, thus forming a nuclear transfer construct; culturing said nuclear transfer construct to produce a viable embryo; and transferring said embryo to the oviducts of a female.
115. The method of claim 114, wherein said enucleated egg is a cumulus-free oocyte.
116. The method of claim 114, wherein said enucleated egg is enucleated pre-metaphase II.
117. The method of claim 114, wherein said enucleated egg is enucleated just prior to metaphase II arrest.
118. The method of claim 114, wherein said extrusion comprises: holding said egg with a holding micropipette; partially dissecting the zonal pellucida of said egg with a needle by making a slit near the first polar body of said egg; extruding the first polar body and adjacent cytoplasm containing the meiotic spindle, ranging from about telophase-I to about pro-metaphase-II, by squeezing said needle.
119. The method of claim 118, wherein said holding micropipette has an about 110 μm innter diameter.
120. The method of claim 118, wherein said needle is a glass needle.
121. The method of claim 114, wherein said egg is enucleated in Hepes-buffered TALP supplemented with BSA and cytochalasin B.
122. The method of claim 114, wherein said egg is enucleated in Hepes-buffered TALP supplemented with about 0.3% BSA and about 7.5 μg/ml cytochalasin B.
123. The method of claim 114, wherein said nuclei are derived from a somatic cell nuclear donor source.
124. The method of claim 123, wherein said nuclei derived from said somatic cell nuclear donor source include dissociated cumulus cells.
125. The method of claim 124, wherein said dissociated cumulus cells are autologous.
126. The method of claim 124, wherein said dissociated cumulus cells are heterologous.
127. The method of claim 124, wherein said dissociated cumulus cells are autologous and heterologous.
128. The method of claim 114, wherein said nuclei are derived from primary rhesus fibroblast cell lines.
129. The method of claim 114, wherein said nuclei are derived from donor blastomeres.
130. The method of claim 114, wherein said nuclei are transferred into the perivitelline space of said enucleated egg.
131. The method of claim 114, wherein said nuclear transfer constructs are equilibrated with mannitol solution.
132. The method of claim 131, wherein said mannitol solution comprises about 0.3 M mannitol solution containing about 0.5 mM Hepes, about 0.1 mM CaCl2, and about 0.1 mM MgCl2
133. The method of claim 131, wherein said nuclear transfer constructs are equilibrated with said mannitol solution for about 4 minutes.
134. The method of claim 131, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing an electrode overlaid with said mannitol solution.
135. The method of claim 131, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing electrodes overlaid with said mannitol solution.
136. The method of claim 135, wherein said chamber contains two electrodes overlaid with said mannitol solution.
137. The method of claim 114, wherein said nuclei and said egg are fused with two DC pulses.
138. The method of claim 137, wherein said DC pulses constitute about 2.7 kK/cm.
139. The method of claim 137, wherein the duration of said DC pulses is about 15 μs.
140. The method of claim 114, wherein said nuclear transfer construct is developed in culture media.
141. The method of claim 140, wherein said culture media includes Gl, G2, and modified synthetic oviductal fluid (mSOF).
142. The method of claim 140, wherein said nuclear transfer construct is developed in said culture media sequentially.
143. The method of claim 140, wherein said nuclear transfer construct is developed in Gl for about 48 hours after nuclear transfer.
144. The method of claim 140, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours.
145. The method of claim 140, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours and transferred to mSOF around the morula stage until said nuclear transfer construct reaches the blastocyst stage.
146. The method of claim 140, wherein said mSOF media further comprises fructose.
147. The method of claim 114, wherein said nuclei have desired characteristics.
148. The method of claim 147, wherein said desired characteristics are linked to a specific disease or disorder.
149. The method of claim 148, wherein said specific disease or disorder is selected from the group consisting of cardiovascular disease, neurological disease, reproductive disorder, cancer, eye disease, endocrine disorder, pulmonary disease, metabolic disorder, autoimmune disorder, and aging.
150. The method of claim 114, wherein said introducing step comprises performing SCNT.
151. The method of claim 150, further comprising the step of performing pronuclear removal after SCNT.
152. The method of claim 150, further comprising the step of performing a second nuclear transfer following said SCNT.
153. The method of claim 114, wherein said introducing step further comprises performing meiotic spindle collapse.
154. The method of claim 114, further comprising the step of performing ooplasmic supplementation following said introducing step.
155. The method of claim 154, wherein said ooplamsic supplementation is performed by ooplast electrofusion.
156. The method of claim 154, wherein said ooplasmic supplementation is performed by microinjection.
157. The method of claim 114, wherein said molecular components comprise centrosomal components normally present in sperm centrosomes.
158. The method of claim 114, wherein said molecular components comprise mitotic motor proteins and centrosome proteins.
159. The method of claim 158, wherein said mitotic motor proteins comprise kinesins.
160. The method of claim 159, wherein said kinesins comprise HSET kinesin.
161. The method of claim 158, wherein said centrosome proteins comprise NuMA.
162. The method of claim 114, wherein said viable embryo is transgenic.
163. The method of claim 162, further comprising the step of producing embryonic stem cells from said viable embryo.
164. The method of claim 163, wherein said embryonic stem cells are human and said viable embryo is human.
165. An embryo produced by the method of claim 114.
166. A method comprising the steps of: introducing nuclei along with one or more molecular components into an extrusion- enucleated egg, thus forming a nuclear transfer construct; culturing said nuclear transfer construct to produce a viable embryo; and dissociating blastomeres from said embryo.
167. The method of claim 166, wherein said enucleated egg is a cumulus-free oocyte.
168. The method of claim 166, wherein said enucleated egg is enucleated pre-metaphase II.
169. The method of claim 166, wherein said enucleated egg is enucleated just prior to metaphase II arrest.
170. The method of claim 166, wherein said extrusion comprises: holding said egg with a holding micropipette; partially dissecting the zonal pellucida of said egg with a needle by making a slit near the first polar body of said egg; extruding the first polar body and adjacent cytoplasm containing the meiotic spindle, ranging from about telophase-I to about pro-metaphase-II, by squeezing said needle.
171. The method of claim 170, wherein said holding micropipette has an about 110 μm inner diameter.
172. The method of claim 170, wherein said needle is a glass needle.
173. The method of claim 166, wherein said egg is enucleated in Hepes-buffered TALP supplemented with BSA and cytochalasin B.
174. The method of claim 166, wherein said egg is enucleated in Hepes-buffered TALP supplemented with about 0.3% BSA and about 7.5 μg/ml cytochalasin B.
175. The method of claim 166, wherein said nuclei are derived from a somatic cell nuclear donor source.
176. The method of claim 175, wherein said nuclei derived from said somatic cell nuclear donor source include dissociated cumulus cells.
177. The method of claim 176, wherein said dissociated cumulus cells are autologous.
178. The method of claim 176, wherein said dissociated cumulus cells are heterologous.
179. The method of claim 176, wherein said dissociated cumulus cells are autologous and heterologous.
180. The method of claim 166, wherein said nuclei are derived from primary rhesus fibroblast cell lines.
181. The method of claim 166, wherein said nuclei are derived from donor blastomeres.
182. The method of claim 166, wherein said nuclei are transferred into the perivitelline space of said enucleated egg.
183. The method of claim 166, wherein said nuclear transfer constructs are equilibrated with mannitol solution.
184. The method of claim 183, wherein said mannitol solution comprises about 0.3 M mannitol solution containing about 0.5 mM Hepes, about 0.1 mM CaCl2, and about 0.1 mM MgCl2
185. The method of claim 183, wherein said nuclear transfer constructs are equilibrated with said mannitol solution for about 4 minutes.
186. The method of claim 183, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing an electrode overlaid with said mannitol solution.
187. The method of claim 183, wherein after said equilibration with said mannitol solution, said nuclear transfer constructs are transferred to a chamber containing electrodes overlaid with said mannitol solution.
188. The method of claim 187, wherein said chamber contains two electrodes overlaid with said mannitol solution.
189. The method of claim 166, wherein said nuclei and said egg are fused with two DC pulses.
190. The method of claim 189, wherein said DC pulses constitute about 2.7 kK/cm.
191. The method of claim 189, wherein the duration of said DC pulses is about 15 μs.
192. The method of claim 166, wherein said nuclear transfer construct is developed in culture media.
193. The method of claim 192, wherein said culture media includes Gl, G2, and modified synthetic oviductal fluid (mSOF).
194. The method of claim 192, wherein said nuclear transfer construct is developed in said culture media sequentially.
195. The method of claim 192, wherein said nuclear transfer construct is developed in Gl for about 48 hours after nuclear transfer.
196. The method of claim 192, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours.
197. The method of claim 192, wherein said nuclear transfer construct is developed in Gl media for about 48 hours after nuclear transfer and then developed in G2 media for an about another 48 hours and transferred to mSOF around the morula stage until said nuclear transfer construct reaches the blastocyst stage.
198. The method of claim 192, wherein said mSOF media further comprises fructose.
199. The method of claim 166, wherein said nuclei have desired characteristics.
200. The method of claim 199, wherein said desired characteristics are linked to a specific disease or disorder.
201. The method of claim 200, wherein said specific disease or disorder is selected from the group consisting of cardiovascular disease, neurological disease, reproductive disorder, cancer, eye disease, endocrine disorder, pulmonary disease, metabolic disorder, autoimmune disorder, and aging.
202. The method of claim 166, wherein said introducing step comprises performing SCNT.
203. The method of claim 202, further comprising the step of performing pronuclear removal after said SCNT.
204. The method of claim 202, further comprising the step of performing a second nuclear transfer following said SCNT.
205. The method of claim 166, prior to said introducing step performing meiotic spindle collapse.
206. The method of claim 166, further comprising the step of performing ooplasmic supplementation following said introducing step.
207. The method of claim 206, wherein said ooplamsic supplementation is performed by ooplast electrofusion.
208. The method of claim 206, wherein said ooplasmic supplementation is performed by microinjection.
209. The method of claim 166, wherein said molecular components comprise centrosomal components normally present in sperm centrosomes.
210. The method of claim 166, wherein said molecular components comprise mitotic motor proteins and centrosome proteins.
211. The method of claim 210, wherein said mitotic motor proteins comprise kinesins.
212. The method of claim 211, wherein said kinesins comprise HSET kinesin.
213. The method of claim 210, wherein said centrosome proteins comprise NuMA.
214. The dissociated blastomeres produced by the method of claim 166.
PCT/US2005/043224 2004-12-03 2005-12-01 Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals WO2006071435A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP05852473A EP1828376A4 (en) 2004-12-03 2005-12-01 Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals
JP2007544448A JP2008521438A (en) 2004-12-03 2005-12-01 How to correct mitotic spindle defects and optimize pre-implantation embryo development rates associated with somatic cell nuclear transfer in animals

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/003,006 2004-12-03
US11/003,006 US20060037086A1 (en) 2003-04-09 2004-12-03 Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals

Publications (2)

Publication Number Publication Date
WO2006071435A2 true WO2006071435A2 (en) 2006-07-06
WO2006071435A3 WO2006071435A3 (en) 2007-06-21

Family

ID=36615367

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/043224 WO2006071435A2 (en) 2004-12-03 2005-12-01 Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals

Country Status (5)

Country Link
US (2) US20060037086A1 (en)
EP (1) EP1828376A4 (en)
JP (1) JP2008521438A (en)
KR (1) KR20070101266A (en)
WO (1) WO2006071435A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008085891A1 (en) * 2007-01-05 2008-07-17 Worcester Polytechnic Institute Oocyte spindle-associated factors improve somatic cell cloning

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004091288A2 (en) * 2003-04-09 2004-10-28 Magee-Womens Health Corporation Correcting mitotic spindle defects in somatic cell nuclear transfer
CN108624621B (en) * 2018-01-17 2019-04-12 中国科学院上海生命科学研究院 The preparation method of the somatic cell clone animal of non-human primates
CN113943712B (en) * 2021-12-20 2022-03-22 南京岚轩生物科技有限公司 Electrofusion buffer solution, preparation method thereof and electrofusion method

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5874301A (en) * 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5775425A (en) * 1995-03-29 1998-07-07 Halliburton Energy Services, Inc. Control of fine particulate flowback in subterranean wells
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
US6011197A (en) * 1997-03-06 2000-01-04 Infigen, Inc. Method of cloning bovines using reprogrammed non-embryonic bovine cells
EP1356035B1 (en) * 2000-12-22 2011-03-16 Kyowa Hakko Kirin Co., Ltd. Methods for cloning non-human mammals using reprogrammed donor chromatin or donor cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1828376A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008085891A1 (en) * 2007-01-05 2008-07-17 Worcester Polytechnic Institute Oocyte spindle-associated factors improve somatic cell cloning

Also Published As

Publication number Publication date
US20060037086A1 (en) 2006-02-16
EP1828376A2 (en) 2007-09-05
EP1828376A4 (en) 2010-11-17
US20090007285A1 (en) 2009-01-01
WO2006071435A3 (en) 2007-06-21
KR20070101266A (en) 2007-10-16
JP2008521438A (en) 2008-06-26

Similar Documents

Publication Publication Date Title
Ogura et al. Behaviour of hamster and mouse round spermatid nuclei incorporated into mature oocytes by electrofusion
JP3739652B2 (en) Full-term growth of animals from enucleated oocytes reconstituted with adult somatic cell nuclei
Galli et al. Introduction to cloning by nuclear transplantation
US20030106082A1 (en) Clonal propagation of primate offspring by embryo splitting
US20100242125A1 (en) Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals
Trounson Nuclear transfer in human medicine and animal breeding
US20090007285A1 (en) Methods for correcting mitotic spindle defects and optimizing preimplantation embryonic developmental rates associated with somatic cell nuclear transfer in animals
US20060168671A1 (en) Injection of caprine sperm factor (cSF), phospholipase C zeta (PLCzeta) and adenophostin A as alternative methods of activation during nuclear transfer in the caprine species
AU784371B2 (en) Nuclear transfer with selected donor cells
US7071372B2 (en) Method for cloning animals with targetted genetic alterations by transfer of long-term cultured male or female somatic cell nuclei, comprising artificially-induced genetic alterations, to enucleated recipient cells
US20040148648A1 (en) Method and system for utilizing somatic cell nuclear transfer embryos as cell donors for additional nuclear transfer
JP2005528095A (en) Methods for selecting cell lines for use in nuclear transfer in mammalian species
US20060174358A1 (en) Method for cloning animals with targetted genetic alterations by transfer of long-term cultured male or female somatic cell nuclei, comprising artificially-induced genetic alterations, to enucleated recipient cells
Chesné et al. Cloning in the rabbit: present situation and prospects
Ogura et al. Microinsemination using spermatogenic cells in mammals

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007544448

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005852473

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020077015240

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2005852473

Country of ref document: EP