WO2001019977A9 - Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions - Google Patents

Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions

Info

Publication number
WO2001019977A9
WO2001019977A9 PCT/US2000/025090 US0025090W WO0119977A9 WO 2001019977 A9 WO2001019977 A9 WO 2001019977A9 US 0025090 W US0025090 W US 0025090W WO 0119977 A9 WO0119977 A9 WO 0119977A9
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
embryonic
stem
human
Prior art date
Application number
PCT/US2000/025090
Other languages
French (fr)
Other versions
WO2001019977A1 (en
Inventor
James Robl
Jose Cibelli
Steven L Stice
Original Assignee
Univ Massachusetts
James Robl
Jose Cibelli
Steven L Stice
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 Univ Massachusetts, James Robl, Jose Cibelli, Steven L Stice filed Critical Univ Massachusetts
Priority to MXPA02002744A priority Critical patent/MXPA02002744A/en
Priority to AU77019/00A priority patent/AU7701900A/en
Priority to JP2001523749A priority patent/JP2003525031A/en
Priority to CA002384413A priority patent/CA2384413A1/en
Priority to IL14854700A priority patent/IL148547A0/en
Priority to NZ517609A priority patent/NZ517609A/en
Priority to BR0013999-8A priority patent/BR0013999A/en
Priority to EP00966717A priority patent/EP1214404A4/en
Publication of WO2001019977A1 publication Critical patent/WO2001019977A1/en
Priority to IL148547A priority patent/IL148547A/en
Publication of WO2001019977A9 publication Critical patent/WO2001019977A9/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • 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

Definitions

  • the present invention generally relates to the production of embryonic or
  • the present invention more specifically relates to the production of primate or
  • oocyte e.g., a primate or ungulate oocyte
  • bovine enucleated oocyte in a preferred embodiment a bovine enucleated oocyte.
  • the present invention further relates to the use of the resultant embryonic
  • stem-like cells preferably primate or human embryonic or stem-like cells for
  • transgenic embryonic or transgenic differentiated cells which may also be used for therapy or diagnosis, and for the production of transgenic embryonic or transgenic differentiated cells, cell lines, tissues and
  • invention may themselves be used as nuclear donors in nuclear transplantation or
  • transgenic cloned or chimeric animals transgenic cloned or chimeric animals.
  • preimplantation mouse embryos are well known. (See, e.g., Evans et al., Nature,
  • ES cells can be passaged in an undifferentiated state, provided that a feeder layer
  • ES cells can be used as an in vitro model
  • livestock animals e.g., ungulates, nuclei from like preimplantation livestock
  • livestock animals are highly desirable because they may provide a potential source
  • the reference disclosed culturing of
  • ICM cells as donor nuclei in nuclear transfer procedures, to produce blastocysts
  • the inner cell mass cells from NT units could be used to form an ES cell-like
  • Collas et al taught the use of granulosa cells (adult somatic
  • alkaline phosphatase activity are pluripotent, and have karyotypes which include
  • SCID mouse Also, purported embryonic stem cell lines derived from human or
  • Parkinson's disease is caused by
  • Parkinson's which promises to have broad applicability to treatment of many brain
  • Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a fetal or neonatal animals into the adult brain. Fetal neurons from a
  • tissue obtained from miscarriages is very limited. Moreover, obtaining cells or
  • stem-like undifferentiated cells for use in transplants and cell and gene thera-
  • nucleus of a mammalian or human cell into an enucleated oocyte of a different
  • a non-human primate or human cell involves transplantation of the nucleus of a non-human primate or human cell into an enucleated animal or human oocyte, e.g., an ungulate, human or primate
  • human somatic cell (karyoplast) with an activated or non-activated, enucleated or
  • non-enucleated oocyte of a different species e.g., bovine
  • a different species e.g., bovine
  • activated or unactivated cross-species NT unit which may be cleaved or uncleaved.
  • human cell e.g., a human adult cell into an enucleated non-human primate or
  • human oocyte wherein such cell has been genetically engineered to be incapable
  • genes of the MHC I family e.g., genes of the MHC I family, and in particular Ped genes such
  • antisense DNA encoding a cell death gene such as BAX, Apaf-1, or capsase, or a
  • apoptosis e.g., by introduction of a DNA construct that provides for the
  • genes that inhibit apoptosis e.g., Bcl-2 or Bcl-2 family members
  • a visualizable (e.g., fluorescent tag) marker protein encodes a visualizable (e.g., fluorescent tag) marker protein.
  • protease inhibitors preferably one or more capsase inhibitors, thereby inhibiting
  • primate or human cell into an enucleated animal oocyte e.g., a human, primate or
  • tissue or organ transplantation is therapeutically or diagnostically beneficial.
  • transplantation therapy comprising the usage of isogenic or synegenic cells
  • Such therapies include by way of example treatment
  • transgenic animals e.g., non-human primates, rodents, ungulates, etc.
  • transgenic animals can be
  • polypeptides e.g., therapeutics or nutripharmaceuticals.
  • Figure 1 is a photograph of a nuclear transfer (NT) unit produced by
  • FIGS. 2 to 5 are photographs of embryonic stem-like cells derived from
  • NT unit such as is depicted in Figure 1.
  • the present invention provides a novel method for producing embryonic
  • species or more specifically adult cells or DNA of one species (e.g., human) and
  • stem-like cells and cell colonies may be obtained by transplantation of the nu-
  • cleus of a human cell e.g., an adult differentiated human cell, into an enucleated
  • animal oocyte which is used to produce nuclear transfer (NT) units, the cells of
  • animal or human cell e.g., adult cell, into the enucleated egg of a different animal
  • the NT units used to produce ES-like cells will be cultured to
  • embryonic or stem-like cells refer to cells
  • stem-like cells rather than stem cells because of the manner in which they are typically produced, i.e., by cross-species nuclear transfer. While these
  • these stem-like cells may possess the mitochondria of the
  • epithelial cell obtained from the oral cavity of a human donor, when transferred
  • oocytes in general comprise factors
  • RNAs and/or RNA sequences may comprise material RNAs and/or DNA sequences.
  • bovine oocytes it is reasonable to expect that human cells may be transplanted into
  • oocytes of other non-related species e.g., other ungulates as well as other animals.
  • ungulate oocytes should be suitable, e.g. pigs, sheep, horses,
  • oocytes from other sources should be suitable, e.g. oocytes
  • the present invention involves the
  • an oocyte (preferably enucleated) of an animal species different from the donor
  • nuclei by injection or fusion, to produce an NT unit containing cells which may
  • the invention may involve the transplantation of an
  • nuclei are combined to produce NT units and which are cultured under conditions suitable to obtain multicellular NT units, preferably comprising at least about 2 to
  • 400 cells more preferably 4 to 128 cells, and most preferably at least about 50
  • the cells of such NT units may be used to produce embryonic or stem-like cells.
  • the preferred embodiment of the invention comprises the
  • enucleated human, primate, or non-primate animal oocyte e.g., an ungulate
  • oocyte and in a preferred embodiment a bovine enucleated oocyte.
  • the embryonic or stem-like cells will be produced by a nuclear radical
  • oocytes obtained from a suitable source, e.g. a mammal and most
  • a primate or an ungulate source e.g. bovine
  • oocyte of an animal species different than the donor cell or nuclei e.g., by fusion
  • steps (iii) and (iv) may be effected in either order;
  • embryonic structures embryoid structures having a discernible inner cell mass
  • Nuclear transfer techniques or nuclear transplantation techniques are examples of nuclear transfer techniques.
  • Human or animal cells preferably mammalian cells, may be obtained and
  • inventions include, by way of example, epithelial, neural cells, epidermal cells,
  • keratinocytes hematopoietic cells, melanocytes, chondrocytes, lymphocytes
  • T lymphocytes other immune cells, erythrocytes, macrophages, melanocytes,
  • monocytes monocytes, mononuclear cells, fibroblasts, cardiac muscle cells, and other muscle
  • the human cells used for nuclear transfer may be obtained.
  • organs e.g., skin, lung, pancreas, liver, stomach, intestine, heart,
  • Suitable donor cells i.e., cells useful in the subject invention, may be obtained from any cell or organ of the body. This
  • the donor cells or nucleus would include all somatic or germ cells.
  • the donor cells or nucleus would include all somatic or germ cells.
  • the donor cells or nucleus would include all somatic or germ cells.
  • the donor cells or nucleus would include all somatic or germ cells.
  • such donor cells will be in the Gl cell
  • the resultant blastocysts may be used to obtain embryonic stem cell lines
  • epithelial cells were epithelial cells derived from the oral cavity of a human donor and adult
  • cell nuclei may be obtained from
  • Zygote gene activation is associated with hyperacetylation of Histone H4.
  • Trichostatin-A has been shown to inhibit histone deacetylase in a reversible
  • Trichostatin A and trapoxin novel chemical probes for the
  • deacetylase Generally, butyrate appears to modify gene expression and in almost
  • donor cells may be exposed to Trichostatin-A or
  • azacytidine can be used to reduce the level of DNA methylation in cells
  • donor cells may be exposed to 5-azacytidine (5-Aza)
  • the oocytes used for nuclear transfer may be obtained from animals
  • Suitable mammalian sources for oocytes including mammals and amphibians. Suitable mammalian sources for oocytes
  • mice include sheep, bovines, ovines, pigs, horses, rabbits, goats, guinea pigs, mice,
  • the oocytes are hamsters, rats, primates, humans, etc.
  • the oocytes are hamsters, rats, primates, humans, etc.
  • the oocytes are hamsters, rats, primates, humans, etc.
  • primates or ungulates e.g., a bovine.
  • oocytes from the ovaries or reproductive tract of a mammal
  • bovine oocytes e.g., bovine oocytes
  • oocytes must generally be matured in vitro before these cells
  • recipient cells may be used as recipient cells for nuclear transfer, and before they can be fertilized
  • immature (prophase I) oocytes from animal ovaries, e.g., bovine ovaries obtained at a slaughterhouse and maturing the oocytes in a maturation medium
  • bovine oocytes generally occurs about 18-24 hours post-
  • this period of time is known as
  • tion refers to aspiration of the immature oocyte from ovarian follicles.
  • metaphase II stage oocytes which have been matured in vivo
  • metaphase II oocytes are collected surgically from either non- superovulated or
  • hCG human chorionic gonadotropin
  • the recipient oocyte because at this stage it is believed that the oocyte can be or is
  • the oocyte activation period In domestic animals, and especially cattle, the oocyte activation period
  • immature oocytes may be washed in HEPES buffered hamster
  • HECM embryo culture medium
  • TCM tissue culture medium
  • FSH follicle stimulating hormone
  • the oocytes will typically be
  • the oocytes Prior to enucleation the oocytes will preferably be removed and
  • enucleation may be effected before or after introduction of donor
  • Enucleation may be effected by known methods, such as described in U.S.
  • Patent No. 4,994,384 which is incorporated by reference herein.
  • metaphase II oocytes are either placed in HECM, optionally containing 7.5 micrograms per milliliter cytochalasin B, for immediate enucleation, or may be
  • a suitable medium for example CRlaa, plus 10% estrus cow serum, and
  • Enucleation may be accomplished microsurgically using a micropipette to
  • the oocytes may then be
  • screening may be effected by staining the oocytes with 1 microgram per milliliter
  • enucleated can then be placed in a suitable culture medium.
  • the recipient oocytes will typically be enucleated
  • vitro maturation more preferably from about 16 hours to about 24 hours after
  • Enucleation may be effected before, simultaneous or
  • enucleation may be effected before, after or
  • perivitelline space of the oocyte typically enucleated, used to produce the NT unit.
  • removal of endogenous nucleus may alternatively be effected after
  • the cells may be fused by electrofusion. Electrofusion is accomplished
  • electrofusion media can be used including e.g., sucrose, mannitol, sorbitol and
  • Fusion can also be accomplished using Sendai virus
  • the human or animal cell and oocyte are electrofused in a 500 .
  • fused NT units are preferably placed in a suitable medium until activation, e.g.,
  • activation may be effected from about twelve hours prior to nuclear
  • activation is effected simultaneous or shortly after nuclear transfer, e.g., about four
  • the NT unit may be activated by known methods. Such methods include,
  • activation may be achieved by application of known
  • activation agents For example, penetration of oocytes by sperm during
  • Suitable oocytes may transfer treatments such as electrical and chemical shock or cycloheximide treatment. Also, treatments such as electrical and chemical shock or cycloheximide treatment may also be used to activate NT embryos after fusion. Suitable oocytes
  • oocyte activation may be effected by simultaneously or
  • oocyte cytoplasm e.g., magnesium, strontium, barium or calcium, e.g., in the form
  • Phosphorylation may be reduced by known methods, e.g., by the addition
  • kinase inhibitors e.g., serine-threonine kinase inhibitors, such as 6-dimethyl-
  • phosphorylation of cellular proteins may be inhibited by
  • a phosphatase into the oocyte, e.g., phosphatase 2A and
  • mice mice, rats, rabbits or hamsters
  • transfer procedures e.g., those including the use of primate or human donor cells
  • both the donor cell and nucleus is of ungulate origin, e.g., a sheep, buffalo, horse,
  • oocyte is of ungulate origin, e.g., sheet, pig,
  • activation may be effected before, simultaneous, or after nuclear
  • oocyte e.g., approximately simultaneous or within about 40 hours of maturation
  • Activated NT units may be cultured in a suitable in vitro culture medium
  • FCS fetal calf serum
  • TCM-199 Medium- 199 + 10% fetal calf serum, Tyrodes-Albumin-Lactate- Pyruvate (TALP), Dulbecco's Phosphate Buffered Saline (PBS), Eagle's and
  • TCM-199 maturation of oocytes is TCM-199, and 1 to 20% serum supplement including fetal
  • preferred maintenance medium includes TCM-199 with Earl salts, 10% fetal calf
  • human epithelial cells of the endometrium secrete leukemia
  • LIF inhibitory factor
  • CR1 contains the nutritional substances necessary to support an
  • CR1 contains hemicalcium L-lactate in amounts ranging from 1.0 mM
  • Hemicalcium L-lactate is L-lactate with
  • hemicalcium salt incorporated thereon.
  • the cultured NT unit or units are preferably washed and then
  • a suitable media e.g., CRIaa medium, Ham's F-10, Tissue Culture Media
  • TCM-199 Tyrodes-Albumin-Lactate-Pyruvate (TALP) Dulbecco's
  • PBS Phosphate Buffered Saline
  • Eagle's or Whitten's preferably containing
  • Suitable feeder layers include, by way
  • fibroblasts and epithelial cells e.g., fibroblasts and uterine epithelial cells
  • the feeder cells will comprise mouse
  • the NT units are cultured on the feeder layer until the NT units reach a size
  • these NT units will be cultured until they reach
  • Another preferred medium comprises ACM + uridine + glucose + 1000 IU
  • the cells used in the present invention will preferably comprise mammalian somatic cells, most preferably cells derived from
  • the donor cell will be genetically modified by the addition,
  • the donor cell For example, the donor cell, the donor cell, the donor cell, and
  • a keratinocyte or fibroblast e.g., of human, primate or bovine origin, may be
  • a desired gene product e.g., therapeutic polypeptide.
  • a desired gene product e.g., therapeutic polypeptide.
  • therapeutic polypeptide examples thereof include
  • lymphokines e.g., IGF-I, IGF-II, interferons, colony stimulating factors,
  • connective tissue polypeptides such as collagens, genetic factors, clotting factors,
  • the donor cells may be modified prior to nuclear
  • One aspect of the invention will involve genetic modification of the donor
  • a human cell e.g., a human cell, such that it is lineage deficient and therefore when used for
  • cells may be genetically modified such that when used as nuclear
  • Examples thereof include:
  • Endoderm GATA-6, GATA-4;
  • Ectoderm RNA helicase A, H beta 58.
  • a desired somatic cell e.g., a human keratinocyte, epithelial cell
  • fibroblast will be genetically engineered such that one or more genes specific
  • This genetically modified cell will be used to produce a lineage-defective
  • nuclear transfer embryo i.e., that does not develop at least one of a functional
  • lineage deficient donor cells may also be genetically modified to
  • the genetically modified donor cell will give rise to a lineage-
  • the donor cell can be modified such that it is “mortal”. This
  • telomere telomerase genes can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozyme telomerase genes. This can be achieved by expressing anti-sense or ribozy
  • blastocysts and ES cells resulting from nuclear transfer may have impaired
  • expression construct can be constructed containing a strong constitutive
  • Suitable selectable markers e.g,. neomycin, ADA, DHFR, and a poly-A
  • sequence e.g., bGH polyA sequence. Also, it may be advantageous to further
  • genes are highly conserved in different species, e.g., bovines, goats, porcine, dogs,
  • donor cells can be engineered to affect other genes that enhance embryonic development.
  • modified donor cells should produce blastocysts and preimplantation stage
  • Still another aspect of the invention involves the construction of donor cells
  • apoptosis examples include, e.g., Bad, Bok, BH3, Bik, Hrk, BNIP3, Bim L , Bad,
  • cell death include, by way of example, BcL-XL, Bcl-w, Mcl-l, Al, Nr-13, BHRF-
  • donor cells can be constructed wherein genes that induce apoptosis
  • apoptosis is enhanced or turned on during embryonic development.
  • this can be effected by introducing a DNA construct that
  • a DNA construct containing a Bcl-2 gene operably
  • a regulatable or constitutive promoter e.g., PGK, SV40, CMV, ubiquitin, or beta-actin, an IRES, a suitable selectable marker, and a poly-A sequence
  • a transgene may be
  • donor cells may be constructed containing
  • Another means of enhancing cloning efficiency is to select cells of a
  • a particular cyclin DNA may be operably linked to a regulatory
  • GFP green fluorescent protein
  • An example thereof is the cyclin Dl gene in order to select for cells that
  • Cyclins are proteins that are expressed only during specific stages of the
  • donor cells will be constructed that express one or
  • cyclin DNA may be operably linked to a regulatory sequence, together with a
  • detectable marker e.g., green fluorescent protein (GFP)
  • GFP green fluorescent protein
  • destruction box and optionally insulation sequences to enhance cyclin and/or
  • cyclin Dl gene which can be used to select for cells that are in Gl.
  • any cyclin gene should be suitable for use in the claimed invention.
  • proteases such as capsases
  • capsase-4 inhibitor I examples include by way of example capsase-4 inhibitor I, capsase-3 inhibitor I, capsase-6
  • NT unit typically will contain at least about 50 cells
  • a feeder layer e.g., irradiated fibroblast cells.
  • the cells used to obtain the stem-like cells or cell colonies will be
  • NT units of smaller or greater cell numbers as
  • oocyte's cytosol may facilitate the dedifferentiation process. This can be
  • cell may be fused with an enucleated oocyte and four to six hours later, without
  • the cells are maintained in the feeder layer in a suitable growth medium
  • alpha MEM e.g., alpha MEM supplemented with 10% FCS and 0.1 mM beta-mercaptoethanol (Sigma) and L-glutamine.
  • FCS fetal calf serum
  • beta-mercaptoethanol Sigma
  • L-glutamine L-glutamine
  • the embryonic or stem-like cells and cell colonies obtained will typically be obtained.
  • the nuclear cell donor rather than the species of the donor oocyte.
  • the nuclear cell donor rather than the species of the donor oocyte.
  • nuclear donor cell into an enucleated bovine oocyte the cells exhibit a morphology
  • bovine ES-like cells more similar to mouse embryonic stem cells than bovine ES-like cells.
  • the individual cells of the human ES-line cell colony are the individual cells of the human ES-line cell colony.
  • the cell colony has a longer cell doubling time, about twice
  • mouse ES cells that of mouse ES cells. Also, unlike bovine and porcine derived ES cells, the
  • primate ES cells produced according to the present methods will exhibit similar characteristics
  • inventions are pluripotent will be confirmed by injecting such cells into an animal,
  • tissue samples e.g., a SCID mouse, or large agricultural animal, and thereafter obtaining tissues
  • mesoderm i.e., mesoderm, ectoderm
  • differentiated tissues i.e., mesoderm, ectoderm
  • the resultant embryonic or stem-like cells and cell lines preferably human
  • embryonic or stem-like cells and cell lines have numerous therapeutic and
  • embryonic or stem-like cells may be used as diagnostic applications. Most especially, such embryonic or stem-like cells.
  • Still another object of the present invention is to improve the efficacy of
  • nuclear transfer e.g., cross-species nuclear transfer by introducing mitochondrial
  • the donor cell is human, human mitochondrial
  • DNA will be derived from cells of the particular donor, e.g., liver cells and tissue.
  • Mitochondria can be isolated from cells in tissue culture, or from tissue.
  • Examples of cells or tissues that may be used as sources of mitochondria include
  • fibroblasts epithelium, liver, lung, keratinocyte, stomach, heart, bladder, pancreas,
  • lymphocytes esophageal, lymphocytes, monocytes, mononuclear cells, cumulus cells, uterine
  • mitochondria can be isolated from tissue culture cells and rat
  • mitochondria comprises human liver tissue because such cells contain a large
  • the isolated DNA can also be further purified, if desired, known methods, e.g., density
  • mouse embryonic stem (ES) cells are known that mouse embryonic stem (ES) cells are known.
  • hematopoietic stem cells capable of differentiating into almost any cell type, e.g., hematopoietic stem cells.
  • the embryonic or stem ⁇ should possess similar differentiation capacity.
  • the embryonic or stem ⁇ should possess similar differentiation capacity.
  • the subject human has a desired cell types according to known methods.
  • the subject human has a desired cell types according to known methods.
  • the subject human has a desired cell types according to known methods.
  • the subject human has a desired cell types according to known methods.
  • the subject human has a desired cell types according to known methods.
  • the subject human has a desired cell types according to known methods.
  • the subject human has a desired cell types according to known methods.
  • embryonic or stem-like cells may be induced to differentiate into hematopoietic
  • stem cells muscle cells, cardiac muscle cells, liver cells, cartilage cells, epithelial cells
  • hematopoietic cells including hematopoietic cells, muscle, cardiac muscle, nerve cells, among others.
  • embryonic stem cells are incorporated by reference in their entirety herein.
  • neural cells e.g., neural cells, muscle cells, hematopoietic cells, etc.
  • muscle cells e.g., hematopoietic cells, etc.
  • hematopoietic cells e.g., neural cells, muscle cells, hematopoietic cells, etc.
  • inducible Bcl-2 or Bcl-xl might be useful for enhancing in vitro development
  • Bcl-2 prevents many, but not all, forms of
  • the subject embryonic or stem-like cells may be used to obtain any desired
  • differentiated cell type therapeutic usages of such differentiated human cells are unparalleled.
  • human hematopoietic stem cells may be used in
  • Hematopoietic stem cells can be obtained, e.g., by fusing adult somatic cells of
  • a cancer or AIDS patient e.g., epithelial cells or lymphocytes with an enucleated
  • oocyte e.g., bovine oocyte
  • embryonic or stem-like cells as described
  • hematopoietic stem cells are obtained. Such hematopoietic cells may be used in
  • disorder may be fused with an enucleated animal oocyte, e.g., a primate or bovine
  • oocyte human embryonic or stem-like cells obtained therefrom, and such cells
  • Parkinson's disease Alzheimer's disease
  • ALS cerebral palsy
  • donor cells may be any suitable differentiated cells.
  • donor cells may be any suitable differentiated cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Psychology (AREA)
  • Biotechnology (AREA)
  • Hematology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Endocrinology (AREA)
  • Hospice & Palliative Care (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)
  • Psychiatry (AREA)
  • Oncology (AREA)
  • Urology & Nephrology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Communicable Diseases (AREA)

Abstract

An improved method of nuclear transfer involving the transplantation of differentiated donor cell nuclei into enucleated oocytes of a species different from the donor cell is provided. The resultant nuclear transfer units are useful for the production of isogenic embryonic stem cells, in particular human isogenic embryonic or stem cells. These embryonic or stem-like cells are useful for producing desired differentiated cells and for introduction, removal or modification, of desired genes, e.g., at specific sites of the genome of such cells by homologous recombination. These cells, which may contain a heterologous gene, are especially useful in cell transplantation therapied and for in vitro study of cell differentiation. Also, methods for improving nuclear transfer efficiency by genetically altering donor cells to inhibit apoptosis, select for a specific cell cycle and/or enhance embryonic growth and development are provided.

Description

EMBRYONIC OR STEM-LIKE CELL LINES PRODUCED BY
CROSS SPECIES NUCLEAR TRANSPLANTATION AND
METHODS FOR ENHANCING EMBRYONIC DEVELOPMENT
BY GENETIC ALTERATION OF DONOR CELLS OR
BY TISSUE CULTURE CONDITIONS
CROSS-REFERENCE TO RELATES APPLICATIONS
This application claims priority under 35 U.S.C. §119 to PCT/US99/04608,
filed on March 2, 1999. Also, this application is a continuation-in-part of U.S.
Serial No. 09/032,995, filed March 2, 1998, which is in turn a continuation-in-part
of U.S. Serial No. 08/699,040, filed on August 19, 1996. All of these applications
are incorporated by reference in their entirety herein.
FIELD OF THE INVENTION
The present invention generally relates to the production of embryonic or
stem-like cells by the transplantation of cell nuclei derived from animal or human
cells into enucleated animal oocytes of a species different from the donor nuclei.
The present invention more specifically relates to the production of primate or
human embryonic or stem-like cells by transplantation of the nucleus of a primate
or human cell into an enucleated animal oocyte, e.g., a primate or ungulate oocyte
and in a preferred embodiment a bovine enucleated oocyte.
The present invention further relates to the use of the resultant embryonic
or stem-like cells, preferably primate or human embryonic or stem-like cells for
therapy, for diagnostic applications, for the production of differentiated cells
which may also be used for therapy or diagnosis, and for the production of transgenic embryonic or transgenic differentiated cells, cell lines, tissues and
organs. Also, the embryonic or stem-like cells obtained according to the present
invention may themselves be used as nuclear donors in nuclear transplantation or
nuclear transfer methods for the production of chimeras or clones, preferably
transgenic cloned or chimeric animals.
BACKGROUND OF THE INVENTION
Methods for deriving embryonic stem (ES) cell lines in vitro from early
preimplantation mouse embryos are well known. (See, e.g., Evans et al., Nature,
29:154-156 (1981); Martin, Proc. Natl. Acad. Set, USA, 78:7634-7638 (1981)).
ES cells can be passaged in an undifferentiated state, provided that a feeder layer
of fibroblast cells (Evans et al., Id) or a differentiation inhibiting source (Smith
et a\., Dev. BioL, 121 : 1-9 (1987)) is present.
ES cells have been previously reported to possess numerous applications.
For example, it has been reported that ES cells can be used as an in vitro model
for differentiation, especially for the study of genes which are involved in the
regulation of early development. Mouse ES cells can give rise to germline
chimeras when introduced into preimplantation mouse embryos, thus demon¬
strating their pluripotency (Bradley et al., Nature, 309:255-256 (1984)). In view of their ability to transfer their genome to the next generation, ES
cells have potential utility for germline manipulation of livestock animals by using
ES cells with or without a desired genetic modification. Moreover, in the case of
livestock animals, e.g., ungulates, nuclei from like preimplantation livestock
embryos support the development of enucleated oocytes to term (Smith et al., Biol.
Reprod., 40:1027-1035 (1989); and Keefer et al., Biol. Reprod., 50:935-939
(1994)). This is in contrast to nuclei from mouse embryos which beyond the eight-
cell stage after transfer reportedly do not support the development of enucleated
oocytes (Cheong et al, Biol. Reprod., 48:958 (1993)). Therefore, ES cells from
livestock animals are highly desirable because they may provide a potential source
of totipotent donor nuclei, genetically manipulated or otherwise, for nuclear trans¬
fer procedures.
Some research groups have reported the isolation of purportedly pluripotent
embryonic cell lines. For example, Notarianni et al., J. Reprod. Fert. Suppl,
43:255-260 (1991), report the establishment of purportedly stable, pluripotent cell
lines from pig and sheep blastocysts which exhibit some morphological and
growth characteristics similar to that of cells in primary cultures of inner cell
masses isolated immunosurgically from sheep blastocysts. (Id.) Also, Notarianni
et al., J. Reprod. Fert. Suppl, 41:51-56 (1990) discloses maintenance and differ-
entiation in culture of putative pluripotential embryonic cell lines from pig blasto¬
cysts. Further, Gerfen et al., Anim. Biotech, 6(1): 1-14 (1995) disclose the isolation of embryonic cell lines from porcine blastocysts. These cells are stably maintained
in mouse embryonic fibroblast feeder layers without the use of conditioned
medium. These cells reportedly differentiate into several different cell types
during culture (Gerfen et al., Id).
Further, Saito et al., Roux's Arch. Dev. Biol, 201:134-141 (1992) report
bovine embryonic stem cell-like cell lines cultured which survived passages for
three, but were lost after the fourth passage. Still further, Handyside et al., Roux's
Arch. Dev. Biol, 196:185-190 (1987) disclose culturing of immunosurgically
isolated inner cell masses of sheep embryos under conditions which allow for the
isolation of mouse ES cell lines derived from mouse ICMs. Handyside et al.
(1987) (Id), report that under such conditions, the sheep ICMs attach, spread, and
develop areas of both ES cell-like and endoderm-like cells, but that after pro¬
longed culture only endoderm-like cells are evident. (Id)
Recently, Cherny et al., Theriogenology, 41:175 (1994) reported
purportedly pluripotent bovine primordial germ cell-derived cell lines maintained
in long-term culture. These cells, after approximately seven days in culture,
produced ES-like colonies which stain positive for alkaline phosphatase (AP),
exhibited the ability to form embryoid bodies, and spontaneously differentiated
into at least two different cell types. These cells also reportedly expressed mRNA
for the transcription factors OCT4, OCT6 and HES 1 , a pattern of homeobox genes
which is believed to be expressed by ES cells exclusively. Also recently, Campbell et al., Nature, 380:64-68 (1996) reported the
production of live lambs following nuclear transfer of cultured embryonic disc
(ED) cells from day nine ovine embryos cultured under conditions which promote
the isolation of ES cell lines in the mouse. The authors concluded based on their
results that ED cells from day nine ovine embryos are totipotent by nuclear transfer
and that totipotency is maintained in culture.
Van Stekelenburg-Hamers et al., Mol. Reprod. Dev., 40:444-454 (1995),
reported the isolation and characterization of purportedly permanent cell lines
from inner cell mass cells of bovine blastocysts. The authors isolated and cultured
ICMs from 8 or 9 day bovine blastocysts under different conditions to determine
which feeder cells and culture media are most efficient in supporting the attach¬
ment and outgrowth of bovine ICM cells. They concluded based on then results
that the attachment and outgrowth of cultured ICM cells is enhanced by the use of
STO (mouse fibroblast) feeder cells (instead of bovine uterus epithelial cells) and
by the use of charcoal-stripped serum (rather than normal serum) to supplement
the culture medium. Van Stekelenburg et al reported, however, that their cell lines
resembled epithelial cells more than pluripotent ICM cells. (Id)
Still further, Smith et al., WO 94/24274, published October 27, 1994,
Evans et al, WO 90/03432, published April 5, 1990, and Wheeler et al, WO
94/26889, published November 24, 1994, report the isolation, selection and propa¬
gation of animal stem cells which purportedly may be used to obtain transgenic animals. Also, Evans et al., WO 90/03432, published on April 5, 1990, reported
the derivation of purportedly pluripotent embryonic stem cells derived from
porcine and bovine species which assertedly are useful for the production of
transgenic animals. Further, Wheeler et al, WO 94/26884, published
November 24, 1994, disclosed embryonic stem cells which are assertedly useful
for the manufacture of chimeric and transgenic ungulates. Thus, based on the
foregoing, it is evident that many groups have attempted to produce ES cell lines,
e.g., because of their potential application in the production of cloned or transgenic
embryos and in nuclear transplantation.
The use of ungulate ICM cells for nuclear transplantation has also been
reported. For example, Collas et al., Mol. Reprod. Dev., 38:264-267 (1994)
disclose nuclear transplantation of bovine ICMs by microinjection of the lysed
donor cells into enucleated mature oocytes. The reference disclosed culturing of
embryos in vitro for seven days to produce fifteen blastocysts which, upon
transferral into bovine recipients, resulted in four pregnancies and two births.
Also, Keefer et al, Biol. Reprod., 50:935-939 (1994), disclose the use of bovine
ICM cells as donor nuclei in nuclear transfer procedures, to produce blastocysts
which, upon transplantation into bovine recipients, resulted in several live
offspring. Further, Sims et al., Proc. Natl. Acad. Set, USA, 90:6143-6147 (1993),
disclosed the production of calves by transfer of nuclei from short-term in vitro
cultured bovine ICM cells into enucleated mature oocytes. Also, the production of live lambs following nuclear transfer of cultured
embryonic disc cells has been reported (Campbell et al., Nature, 380:64-68
(1996)). Still further, the use of bovine pluripotent embryonic cells in nuclear
transfer and the production of chimeric fetuses has also been reported (Stice et al.,
Biol. Reprod., 54:100-110 (1996)); Collas et al, Mol. Reprod. Dev., 38:264-267
(1994).
Further, there have been previous attempts to produce cross species NT
units (Wolfe et al., Theriogenology, 33:350 (1990). Specifically, bovine
embryonic cells were fused with bison oocytes to produce some cross species NT
units possibly having an inner cell mass. However, embryonic cells, not adult cells
were used, as donor nuclei in the nuclear transfer procedure. The dogma has been
that embryonic cells are more easily reprogrammed than adult cells. This dates
back to earlier NT studies in the frog (review by DiBerardino, Differentiation,
17:17-30 (1980)). Also, this study involved very phylogenetically similar animals
(cattle nuclei and bison oocytes). By contrast, previously when more diverse
species were fused during NT (cattle nuclei into hamster oocytes), no inner cell
mass structures were obtained. Further, it has never been previously reported that
the inner cell mass cells from NT units could be used to form an ES cell-like
colony that could be propagated.
Also, Collas et al (Id), taught the use of granulosa cells (adult somatic
cells) to produce bovine nuclear transfer embryos. However, unlike the present invention, these experiments did not involve cross-species nuclear transfer. Also,
unlike the present invention ES-like cell colonies were not obtained.
Recently, U.S. Patent No. 5,843,780, issued to James A. Thomson on
December 1, 1998, assigned to the Wisconsin Alumni Research Foundation,
purports to disclose a purified preparation of primate embryonic stem cells that are
(i) capable of proliferation in an in vitro culture for over one year; (ii) maintain a
karyotype in which all chromosomes characteristic of the primate species are
present and not noticeably altered through prolonged culture; (iii) maintains the
potential to differentiate into derivatives of endoderm, mesoderm and ectoderm
tissues throughout culture; and (iv) will not differentiate when cultured on a
fibroblast feeder layer. These cells were reportedly negative for the SSEA-1
marker, positive for the SEA-3 marker, positive for the SSEA-4 marker, express
alkaline phosphatase activity, are pluripotent, and have karyotypes which include
the presence of all the chromosomes characteristic of the primate species and in
which none of the chromosomes are altered. Further, these cells are respectfully
positive for the TRA-1-60, and TRA-1-81 markers. The cells purportedly
differentiate into endoderm, mesoderm and ectoderm cells when injected into a
SCID mouse. Also, purported embryonic stem cell lines derived from human or
primate blastocysts are discussed in Thomson et al., Science 282:1145-1147 and
Proc. Natl. Acad. Set, USA 92:7844-7848 (1995). Thus, Thomson disclose what purportedly are non-human primate and
human embryonic or stem-like cells and methods for their production. However,
there still exists a significant need for methods for producing human embryonic
or stem-like cells that are autologous to an intended transplant recipient given their
5 significant therapeutic and diagnostic potential.
In this regard, numerous human diseases have been identified which may
be treated by cell transplantation. For example, Parkinson's disease is caused by
degeneration of dopaminergic neurons in the substantia nigra. Standard treatment
for Parkinson's involves administration of L-DOPA, which temporarily
o ameliorates the loss of dopamine, but causes severe side effects and ultimately
does not reverse the progress of the disease. A different approach to treating
Parkinson's, which promises to have broad applicability to treatment of many brain
diseases and central nervous system injury, involves transplantation of cells or
tissues from fetal or neonatal animals into the adult brain. Fetal neurons from a
5 variety of brain regions can be incorporated into the adult brain. Such grafts have
been shown to alleviate experimentally induced behavioral deficits, including
complex cognitive functions, in laboratory animals. Initial test results from human
clinical trials have also been promising. However, supplies of human fetal cells
or tissue obtained from miscarriages is very limited. Moreover, obtaining cells or
o tissues from aborted fetuses is highly controversial. There is currently no available procedure for producing "fetal-like" cells
from the patient. Further, allograft tissue is not readily available and both allograft
and xenograft tissue are subject to graft rejection. Moreover, in some cases, it
would be beneficial to make genetic modifications in cells or tissues before trans-
plantation. However, many cells or tissues wherein such modification would be
desirable do not divide well in culture and most types of genetic transformation
require rapidly dividing cells.
There is therefore a clear need in the art for a supply of human embryonic
or stem-like undifferentiated cells for use in transplants and cell and gene thera-
pies.
OBJECTS OF THE INVENTION
It is an object of the invention to provide novel and improved methods for
producing embryonic or stem-like cells.
It is a more specific object of the invention to provide a novel method for
producing embryonic or stem-like cells which involves transplantation of the
nucleus of a mammalian or human cell into an enucleated oocyte of a different
species.
It is another specific object of the invention to provide a novel method for
producing non-human primate or human embryonic or stem-like cells which
involves transplantation of the nucleus of a non-human primate or human cell into an enucleated animal or human oocyte, e.g., an ungulate, human or primate
enucleated oocyte.
It is another object of the invention to enhance the efficacy of cross-species
nuclear transfer by incorporating mitochondrial DNA derived from the same
species (preferably same donor) as the donor cell into the oocyte of a different
species that is used for nuclear transfer, before or after enucleation, or into the
nuclear transfer unit (after the donor cell has been introduced).
It is still another object of the invention to enhance the efficacy of cross-
species nuclear transfer by fusing an enucleated somatic cell (e.g., an enucleated
human somatic cell) (karyoplast) with an activated or non-activated, enucleated or
non-enucleated oocyte of a different species, e.g., bovine, or by fusion with an
activated or unactivated cross-species NT unit which may be cleaved or uncleaved.
It is another object of the invention to provide a novel method for
producing lineage-defective non-human primate or human embryonic or stem-like
cells which involves transplantation of the nucleus of a non-human primate or
human cell, e.g., a human adult cell into an enucleated non-human primate or
human oocyte, wherein such cell has been genetically engineered to be incapable
of differentiation into a specific cell lineage or has been modified such that the
cells are "mortal", and thereby do not give rise to a viable offspring, e.g., by
engineering expression of anti-sense or ribozyme telomerase gene. It is still another object of the invention to enhance efficiency of nuclear
transfer and specifically to enhance the development of preimplantation embryos
produced by nuclear transfer by genetically engineering donor somatic cells used
for nuclear transfer to provide for the expression of genes that enhance embryonic
development, e.g., genes of the MHC I family, and in particular Ped genes such
as Q7 and/or Q9.
It is another object of the invention to enhance the production of nuclear
transfer embryos, e.g., cross-species nuclear transfer embryos, by the introduction
of transgenes before or after nuclear transfer that provide for the expression of an
antisense DNA encoding a cell death gene such as BAX, Apaf-1, or capsase, or a
portion thereof, or demethylase.
It is yet another object of the invention to enhance the production of nuclear
transfer embryos by IVP and more specifically nuclear transfer embryos by
genetically altering the donor cell used for nuclear transfer such that it is resistant
to apoptosis, e.g., by introduction of a DNA construct that provides for the
expression of genes that inhibit apoptosis, e.g., Bcl-2 or Bcl-2 family members
and/or by the expression of antisense ribozymes specific to genes that induce
apoptosis during early embryonic development.
It is still another object of the invention to improve the efficacy of nuclear
transfer by improved selection of donor cells of a specific cell cycle stage, e.g., Gl
phase, by genetically engineering donor cells such that they express a DNA construct encoding a particular cyclin linked to a detectable marker, e.g., one that
encodes a visualizable (e.g., fluorescent tag) marker protein.
It is also an object of the invention to enhance the development of in vitro
produced embryos, by culturing such embryos in the presence of one or more
protease inhibitors, preferably one or more capsase inhibitors, thereby inhibiting
apoptosis.
It is another object of the invention to provide embryonic or stem-like cells
produced by transplantation of nucleus of an animal or human cell into an
enucleated oocyte of a different species.
It is a more specific object of the invention to provide primate or human
embryonic or stem-like cells produced by transplantation of the nucleus of a
primate or human cell into an enucleated animal oocyte, e.g., a human, primate or
ungulate enucleated oocyte.
It is another object of the invention to use such embryonic or stem-like cells
for therapy or diagnosis .
It is a specific object of the invention to use such primate or human
embryonic or stem-like cells for treatment or diagnosis of any disease wherein cell,
tissue or organ transplantation is therapeutically or diagnostically beneficial.
It is another specific object of the invention to use the embryonic or stem-
like cells produced according to the invention for the production of differentiated
cells, tissues or organs. It is a more specific object of the invention to use the primate or human
embryonic or stem-like cells produced according to the invention for the
production of differentiated human cells, tissues or organs.
It is another specific object of the invention to use the embryonic or stem-
like cells produced according to the invention for the production of genetically
engineered embryonic or stem-like cells, which cells may be used to produce
genetically engineered or transgenic differentiated human cells, tissues or organs,
e.g., having use in gene therapies.
It is another specific object of the invention to use the embryonic or stem-
like cells produced according to the invention in vitro, e.g. for study of cell dif¬
ferentiation and for assay purposes, e.g. for drug studies.
It is another object of the invention to provide improved methods of
transplantation therapy, comprising the usage of isogenic or synegenic cells,
tissues or organs produced from the embryonic or stem-like cells produced
according to the invention. Such therapies include by way of example treatment
of diseases and injuries including Parkinson's, Huntington's, Alzheimer's, ALS,
spinal cord injuries, multiple sclerosis, muscular dystrophy, diabetes, liver
diseases, heart disease, cartilage replacement, burns, vascular diseases, urinary
tract diseases, as well as for the treatment of immune defects, bone marrow trans-
plantation, cancer, among other diseases. It is another object of the invention to use the transgenic or genetically
engineered embryonic or stem-like cells produced according to the invention for
gene therapy, in particular for the treatment and/or prevention of the diseases and
injuries identified, supra.
It is another object of the invention to use the embryonic or stem-like cells
produced according to the invention or transgenic or genetically engineered
embryonic or stem-like cells produced according to the invention as nuclear
donors for nuclear transplantation.
It is still another object of the invention to use genetically engineered ES
cells produced according to the invention for the production of transgenic animals,
e.g., non-human primates, rodents, ungulates, etc. Such transgenic animals can be
used to produce, e.g., animal models for study of human diseases, or for the
production of desired polypeptides, e.g., therapeutics or nutripharmaceuticals.
With the foregoing and other objects, advantages and features of the inven-
tion that will become hereinafter apparent, the nature of the invention may be more
clearly understood by reference to the following detailed description of the
preferred embodiments of the invention and to the appended claims.
BRIEFS DESCRIPTION OF THE FIGURES
Figure 1 is a photograph of a nuclear transfer (NT) unit produced by
transfer of an adult human cell into an enucleated bovine oocyte. Figures 2 to 5 are photographs of embryonic stem-like cells derived from
a NT unit such as is depicted in Figure 1.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a novel method for producing embryonic
or stem-like cells, and more specifically non-human primate or human embryonic
or stem-like cells by nuclear transfer or nuclear transplantation. In the subject
application, nuclear transfer or nuclear transplantation or NT are used
interchangeably.
As discussed supra, the isolation of actual embryonic or stem- like cells by
nuclear transfer or nuclear transplantation has never been reported. Rather,
previous reported isolation of ES-like cells has been from fertilized embryos.
Also, successful nuclear transfer involving cells or DNA of genetically dissimilar
species, or more specifically adult cells or DNA of one species (e.g., human) and
oocytes of another non-related species has never been reported. Rather, while
embryos produced by fusion of cells of closely related species, has been reported,
e.g., bovine-goat and bovine-bison, they did not produce ES cells. (Wolfe et al,
Theriogenology, 33(1):350 (1990).) Also, there has never been reported a method
for producing primate or human ES cells derived from a non- fetal tissue source.
Rather, the limited human fetal cells and tissues which are currently available
must be obtained or derived from spontaneous abortion tissues and from aborted
fetuses. Also, prior to the present invention, no one obtained embryonic or stem¬
like cells by cross-species nuclear transplantation.
Quite unexpectedly, the present inventors discovered that human embryonic
or stem-like cells and cell colonies may be obtained by transplantation of the nu-
cleus of a human cell, e.g., an adult differentiated human cell, into an enucleated
animal oocyte, which is used to produce nuclear transfer (NT) units, the cells of
which upon culturing give rise to human embryonic or stem-like cells and cell
colonies. This result is highly surprising because it is the first demonstration of
effective cross-species nuclear transplantation involving the introduction of a
differentiated donor cell or nucleus into an enucleated oocyte of a genetically
dissimilar species, e.g., the transplantation of cell nuclei from a differentiated
animal or human cell, e.g., adult cell, into the enucleated egg of a different animal
species, to produce nuclear transfer units containing cells which when cultured
under appropriate conditions give rise to embryonic or stem-like cells and cell
colonies.
Preferably, the NT units used to produce ES-like cells will be cultured to
a size of at least 2 to 400 cells, preferably 4 to 128 cells, and most preferably to a
size of at least about 50 cells.
In the present invention, embryonic or stem-like cells refer to cells
produced according to the present invention. The present application refers to
such cells as stem-like cells rather than stem cells because of the manner in which they are typically produced, i.e., by cross-species nuclear transfer. While these
cells are expected to possess similar differentiation capacity as normal stem cells
they may possess some insignificant differences because of the manner they are
produced. For example, these stem-like cells may possess the mitochondria of the
oocytes used for nuclear transfer, and thus not behave identically to conventional
embryonic stem cells.
The present discovery was made based on the observation that nuclear
transplantation of the nucleus of an adult human cell, specifically a human
epithelial cell obtained from the oral cavity of a human donor, when transferred
into an enucleated bovine oocyte, resulted in the formation of nuclear transfer
units, the cells of which upon culturing gave rise to human stem-like or embryonic
cells and human embryonic or stem-like cell colonies. This result has recently
been reproduced by transplantation of keratinocytes from an adult human into an
enucleated bovine oocyte with the successful production of a blastocyst and ES
cell line. Based thereon, it is hypothesized by the present inventors that bovine
oocytes and human oocytes, and likely mammals in general must undergo matu¬
ration processes during embryonic development which are sufficiently similar or
conserved so as to permit the bovine oocyte to function as an effective substitute
or surrogate for a human oocyte. Apparently, oocytes in general comprise factors,
likely proteinaceous or nucleic acid in nature, that induce embryonic development
under appropriate conditions, and these functions that are the same or very similar in different species. These factors may comprise material RNAs and/or
telomerase.
Based on the fact that human cell nuclei can be effectively transplanted into
bovine oocytes, it is reasonable to expect that human cells may be transplanted into
oocytes of other non-related species, e.g., other ungulates as well as other animals.
In particular, other ungulate oocytes should be suitable, e.g. pigs, sheep, horses,
goats, etc. Also, oocytes from other sources should be suitable, e.g. oocytes
derived from other primates, amphibians, rodents, rabbits, guinea pigs, etc.
Further, using similar methods, it should be possible to transfer human cells or cell
nuclei into human oocytes and use the resultant blastocysts to produce human ES
cells.
Therefore, in its broadest embodiment, the present invention involves the
transplantation of an animal or human cell nucleus or animal or human cell into
an oocyte (preferably enucleated) of an animal species different from the donor
nuclei, by injection or fusion, to produce an NT unit containing cells which may
be used to obtain embryonic or stem-like cells and/or cell cultures. Enucleation
(removal of endogenous oocyte nucleus) may be effected before or after nuclear
transfer. For example, the invention may involve the transplantation of an
ungulate cell nucleus or ungulate cell into an enucleated oocyte of another species,
e.g., another ungulate or non-ungulate, by injection or fusion, which cells and/or
nuclei are combined to produce NT units and which are cultured under conditions suitable to obtain multicellular NT units, preferably comprising at least about 2 to
400 cells, more preferably 4 to 128 cells, and most preferably at least about 50
cells. The cells of such NT units may be used to produce embryonic or stem-like
cells or cell colonies upon culturing.
However, the preferred embodiment of the invention comprises the
production of non-human primate or human embryonic or stem-like cells by
transplantation of the nucleus of a donor human cell or a human cell into an
enucleated human, primate, or non-primate animal oocyte, e.g., an ungulate
oocyte, and in a preferred embodiment a bovine enucleated oocyte.
In general, the embryonic or stem-like cells will be produced by a nuclear
transfer process comprising the following steps:
(i) obtaining desired human or animal cells to be used as a source of donor
nuclei (which may be genetically altered);
(ii) obtaining oocytes from a suitable source, e.g. a mammal and most
preferably a primate or an ungulate source, e.g. bovine,
(iii) enucleating said oocytes by removal of endogenous nucleus;
(iv) transferring the human or animal cell or nucleus into the enucleated
oocyte of an animal species different than the donor cell or nuclei, e.g., by fusion
or injection, wherein steps (iii) and (iv) may be effected in either order;
(v) culturing the resultant NT product or NT unit to produce multiple cell
structures (embryoid structures having a discernible inner cell mass); and (vi) culturing cells obtained from said embryos to obtain embryonic or
stem-like cells and stem-like cell colonies.
Nuclear transfer techniques or nuclear transplantation techniques are
known in the literature and are described in many of the references cited in the
Background of the Invention. See, in particular, Campbell et al, Theriogenology,
43:181 (1995); Collas et al, Mol. Report Dev., 38:264-267 (1994); Keefer et al,
Biol. Reprod., 50:935-939 (1994); Sims et al, Proc. Natl. Acad. Set, USA,
90:6143-6147 (1993); WO 94/26884; WO 94/24274, and WO 90/03432, which are
incorporated by reference in their entirety herein. Also, U.S. Patent Nos.
4,944,384 and 5,057,420 describe procedures for bovine nuclear transplantation.
See, also Cibelli et al, Science, Vol. 280:1256-1258 (1998).
Human or animal cells, preferably mammalian cells, may be obtained and
cultured by well known methods. Human and animal cells useful in the present
invention include, by way of example, epithelial, neural cells, epidermal cells,
keratinocytes, hematopoietic cells, melanocytes, chondrocytes, lymphocytes (B
and T lymphocytes), other immune cells, erythrocytes, macrophages, melanocytes,
monocytes, mononuclear cells, fibroblasts, cardiac muscle cells, and other muscle
cells, etc. Moreover, the human cells used for nuclear transfer may be obtained
from different organs, e.g., skin, lung, pancreas, liver, stomach, intestine, heart,
reproductive organs, bladder, kidney, urethra and other urinary organs, etc. These
are just examples of suitable donor cells. Suitable donor cells, i.e., cells useful in the subject invention, may be obtained from any cell or organ of the body. This
includes all somatic or germ cells. Preferably, the donor cells or nucleus would
comprise actively dividing, i.e., non-quiescent, cells as this has been reported to
enhance cloning efficacy. Also preferably, such donor cells will be in the Gl cell
cycle.
The resultant blastocysts may be used to obtain embryonic stem cell lines
according to the culturing methods reported by Thomson et al., Science, 282: 1145-
1147 (1998) and Thomson et al., Proc. Natl. Acad. Set, USA, 92:7544-7848
(1995), incorporated by reference in their entirety herein.
In the Example which follows, the cells used as donors for nuclear transfer
were epithelial cells derived from the oral cavity of a human donor and adult
human keratinocytes. However, as discussed, the disclosed method is applicable
to other human cells or nuclei. Moreover, the cell nuclei may be obtained from
both human somatic and germ cells.
It is also possible to arrest donor cells at mitosis before nuclear transfer,
using a suitable technique known in the art. Methods for stopping the cell cycle
at various stages have been thoroughly reviewed in U.S. Patent 5,262,409, which
is herein incorporated by reference. In particular, while cycloheximide has been
reported to have an inhibitory effect on mitosis (Bowen and Wilson (1955) J.
Heredity, 45:3-9), it may also be employed for improved activation of mature bovine follicular oocytes when combined with electric pulse treatment (Yang et
al. (1992) Biol. Reprod., 42 (Suppl. 1): 117).
Zygote gene activation is associated with hyperacetylation of Histone H4.
Trichostatin-A has been shown to inhibit histone deacetylase in a reversible
manner (Adenot et al. Differential H4 acetylation of paternal and maternal
chromatin precedes DNA replication and differential transcriptional activity in
pronuclei of 1-cell mouse embryos. Development (Nov. 1997) 124(22): 4615-
4625; Yoshida et al. Trichostatin A and trapoxin: novel chemical probes for the
role of histone acetylation in chromatin structure and function. Bioessays (May,
1995) 17(5): 423-430), as have other compounds. For instance, butyrate is
also believed to cause hyper-acetylations of histones by inhibiting histone
deacetylase. Generally, butyrate appears to modify gene expression and in almost
all cases its addition to cells in culture appears to arrest cell growth. Use of
butyrate in this regard is described in U.S. Patent No. 5,681,718, which is herein
incorporated by reference. Thus, donor cells may be exposed to Trichostatin-A or
another appropriate deacetylase inhibitor prior to fusion, or such a compound may
be added to the culture media prior to genome activation.
Additionally, demethylation of DNA is thought to be a requirement for
proper access of transcription factors to DNA regulatory sequences. Global
demethylation of DNA from the eight-cell stage to the blastocyst stage in
preimplantation embryos has previously been described (Stein et al., Mol. Reprod. & Dev., 47(4): 421-429). Also, Jaenisch et al. (1997) have reported that 5-
azacytidine can be used to reduce the level of DNA methylation in cells,
potentially leading to increased access of transcription factors to DNA regulatory
sequences. Accordingly, donor cells may be exposed to 5-azacytidine (5-Aza)
previous to fusion, or 5-Aza may be added to the culture medium from the 8 cell
stage to blastocyst. Alternatively, other known methods for effecting DNA
demethylation may be used.
The oocytes used for nuclear transfer may be obtained from animals
including mammals and amphibians. Suitable mammalian sources for oocytes
include sheep, bovines, ovines, pigs, horses, rabbits, goats, guinea pigs, mice,
hamsters, rats, primates, humans, etc. In the preferred embodiments, the oocytes
will be obtained from primates or ungulates, e.g., a bovine.
Methods for isolation of oocytes are well known in the art. Essentially, this
will comprise isolating oocytes from the ovaries or reproductive tract of a mammal
or amphibian, e.g., a bovine. A readily available source of bovine oocytes is
slaughterhouse materials.
For the successful use of techniques such as genetic engineering, nuclear
transfer and cloning, oocytes must generally be matured in vitro before these cells
may be used as recipient cells for nuclear transfer, and before they can be fertilized
by the sperm cell to develop into an embryo. This process generally requires
collecting immature (prophase I) oocytes from animal ovaries, e.g., bovine ovaries obtained at a slaughterhouse and maturing the oocytes in a maturation medium
prior to fertilization or enucleation until the oocyte attains the metaphase II stage,
which in the case of bovine oocytes generally occurs about 18-24 hours post-
aspiration. For purposes of the present invention, this period of time is known as
the "maturation period." As used herein for calculation of time periods, "aspira¬
tion" refers to aspiration of the immature oocyte from ovarian follicles.
Additionally, metaphase II stage oocytes, which have been matured in vivo
have been successfully used in nuclear transfer techniques. Essentially, mature
metaphase II oocytes are collected surgically from either non- superovulated or
superovulated cows or heifers 35 to 48 hours past the onset of estrus or past the
injection of human chorionic gonadotropin (hCG) or similar hormone.
The stage of maturation of the oocyte at enucleation and nuclear transfer
has been reported to be significant to the success of NT methods. (See e.g.,
Prather et al., Differentiation, 48, 1-8, 1991). In general, previous successful
mammalian embryo cloning practices used metaphase II stage oocyte as the
recipient oocyte because at this stage it is believed that the oocyte can be or is
sufficiently "activated" to treat the introduced nucleus as it does a fertilizing
sperm. In domestic animals, and especially cattle, the oocyte activation period
generally ranges from about 16-52 hours, preferably about 28-42 hours post-
aspiration. For example, immature oocytes may be washed in HEPES buffered hamster
embryo culture medium (HECM) as described in Seshagine et al., Biol. Reprod.,
40, 544-606, 1989, and then placed into drops of maturation medium consisting
of 50 microliters of tissue culture medium (TCM) 199 containing 10% fetal calf
serum which contains appropriate gonadotropins such as luteinizing hormone (LH)
and follicle stimulating hormone (FSH), and estradiol under a layer of lightweight
paraffin or silicon at 39°C.
After a fixed time maturation period, which typically will range from about
10 to 40 hours, and preferably about 16-18 hours, the oocytes will typically be
enucleated. Prior to enucleation the oocytes will preferably be removed and
placed in HECM containing 1 milligram per milliliter of hyaluronidase prior to
removal of cumulus cells. This may be effected by repeated pipetting through very
fine bore pipettes or by vortexing briefly. The stripped oocytes are then screened
for polar bodies, and the selected metaphase II oocytes, as determined by the
presence of polar bodies, are then used for nuclear transfer. Enucleation follows.
As noted above, enucleation may be effected before or after introduction of donor
cell or nucleus because the donor nucleus is readily discernible from endogenous
nucleus.
Enucleation may be effected by known methods, such as described in U.S.
Patent No. 4,994,384 which is incorporated by reference herein. For example,
metaphase II oocytes are either placed in HECM, optionally containing 7.5 micrograms per milliliter cytochalasin B, for immediate enucleation, or may be
placed in a suitable medium, for example CRlaa, plus 10% estrus cow serum, and
then enucleated later, preferably not more than 24 hours later, and more preferably
16-18 hours later.
Enucleation may be accomplished microsurgically using a micropipette to
remove the polar body and the adjacent cytoplasm. The oocytes may then be
screened to identify those of which have been successfully enucleated. This
screening may be effected by staining the oocytes with 1 microgram per milliliter
33342 Hoechst dye in HECM, and then viewing the oocytes under ultraviolet
irradiation for less than 10 seconds. The oocytes that have been successfully
enucleated can then be placed in a suitable culture medium.
In the present invention, the recipient oocytes will typically be enucleated
at a time ranging from about 10 hours to about 40 hours after the initiation of in
vitro maturation, more preferably from about 16 hours to about 24 hours after
initiation of in vitro maturation, and most preferably about 16-18 hours after initi¬
ation of in vitro maturation. Enucleation may be effected before, simultaneous or
after nuclear transfer. Also, enucleation may be effected before, after or
simultaneous to activation.
A single animal or human cell or nucleus derived therefrom which is
typically heterologous to the enucleated oocyte will then be transferred into the
perivitelline space of the oocyte, typically enucleated, used to produce the NT unit. However, removal of endogenous nucleus may alternatively be effected after
nuclear transfer. The animal or human cell or nucleus and the enucleated oocyte
will be used to produce NT units according to methods known in the art. For
example, the cells may be fused by electrofusion. Electrofusion is accomplished
by providing a pulse of electricity that is sufficient to cause a transient break down
of the plasma membrane. This breakdown of the plasma membrane is very short
because the membrane reforms rapidly. Essentially, if two adjacent membranes
are induced to break down, upon reformation the lipid bilayers intermingle and
small channels will open between the two cells. Due to the thermodynamic insta-
bility of such a small opening, it enlarges until the two cells become one.
Reference is made to U.S. Patent 4,997,384, by Prather et al., (incorporated by
reference in its entirety herein) for a further discussion of this process. A variety
of electrofusion media can be used including e.g., sucrose, mannitol, sorbitol and
phosphate buffered solution. Fusion can also be accomplished using Sendai virus
as a fusogenic agent (Graham, Wister Inot. Symp. Monogr., 9, 19, 1969).
Also, in some cases (e.g. with small donor nuclei) it may be preferable to
inject the nucleus directly into the oocyte rather than using electroporation fusion.
Such techniques are disclosed in Collas and Barnes, Mol. Reprod. Dev., 38:264-
267 (1994), and incorporated by reference in its entirety herein.
' Preferably, the human or animal cell and oocyte are electrofused in a 500
μm chamber by application of an electrical pulse of 90- 120V for about 15 μsec, about 24 hours after initiation of oocyte maturation. After fusion, the resultant
fused NT units are preferably placed in a suitable medium until activation, e.g.,
one identified infra. Typically activation will be effected shortly thereafter,
typically less than 24 hours later, and preferably about 4-9 hours later. However,
it is also possible to activate the recipient oocyte before or proximate
(simultaneous) to nuclear transfer, which may or may not be enucleated. For
example, activation may be effected from about twelve hours prior to nuclear
transfer to about twenty-four hours after nuclear transfer. More typically,
activation is effected simultaneous or shortly after nuclear transfer, e.g., about four
to nine hours later.
The NT unit may be activated by known methods. Such methods include,
e.g., culturing the NT unit at sub-physiological temperature, in essence by applying
a cold, or actually cool temperature shock to the NT unit. This may be most
conveniently done by culturing the NT unit at room temperature, which is cold
relative to the physiological temperature conditions to which embryos are normally
exposed.
Alternatively, activation may be achieved by application of known
activation agents. For example, penetration of oocytes by sperm during
fertilization has been shown to activate prefusion oocytes to yield greater numbers
of viable pregnancies and multiple genetically identical calves after nuclear
transfer. Also, treatments such as electrical and chemical shock or cycloheximide treatment may also be used to activate NT embryos after fusion. Suitable oocyte
activation methods are the subject of U.S. Patent No. 5,496,720, to Susko-Parrish
et al., which is herein incorporated by reference.
For example, oocyte activation may be effected by simultaneously or
sequentially:
(i) increasing levels of divalent cations in the oocyte, and
(ii) reducing phosphorylation of cellular proteins in the oocyte.
This will generally be effected by introducing divalent cations into the
oocyte cytoplasm, e.g., magnesium, strontium, barium or calcium, e.g., in the form
of an ionophore. Other methods of increasing divalent cation levels include the
use of electric shock, treatment with ethanol and treatment with caged chelators.
Phosphorylation may be reduced by known methods, e.g., by the addition
of kinase inhibitors, e.g., serine-threonine kinase inhibitors, such as 6-dimethyl-
amino-purine, staurosporine, 2-aminopurine, and sphingosine.
Alternatively, phosphorylation of cellular proteins may be inhibited by
introduction of a phosphatase into the oocyte, e.g., phosphatase 2A and
phosphatase 2B.
Specific examples of activation methods are listed below.
1. Activation by Ionomycin and DMAP
1 - Place oocytes in Ionomycin (5 μM) with 2 mM of DMAP for
4 minutes; 2- Move the oocytes into culture media with 2 mM of DMAP
for 4 hours;
3- Rinse four times and place in culture.
2. Activation by Ionomycin DMAP and Roscovitin
1 - Place oocytes in Ionomycin (5 μM) with 2 mM of DMAP for
four minutes;
2- Move the oocytes into culture media with 2 mM of DMAP
and 200 microM of Roscovitin for three hours;
3- Rinse four times and place in culture.
3. Activation by exposure to Ionomycin followed by eytochalasin
and cycloheximide.
1- Place oocytes in Ionomycin (5 microM) for four minutes;
2- Move oocytes to culture media containing 5 μg/ml of
eytochalasin B and 5 μg/ml of cycloheximide for five hours;
3- Rinse four times and place in culture.
4. Activation by electrical pulses
1- Place eggs in mannitol media containing 100 μM CaCL2;
2- Deliver three pulses of 1.0 kVcm"1 for 20 μsec, each pulse 22
minutes apart;
3- Move oocytes to culture media containing 5 μg/ml of
eytochalasin B for three hours. 5. Activation by exposure with ethanol followed by eytochalasin
and cycloheximide
1- Place oocytes in 7% ethanol for one minute;
2- Move oocytes to culture media containing 5 μg/ml of
eytochalasin B and 5 μg/ml of cycloheximide for five hours;
3- Rinse four times and place in culture.
6. Activation by microinjection of adenophostine
1- Inject oocytes with 10 to 12 picoliters of a solution
containing 10 μM of adenophostine;
2- Put oocytes in culture.
7. Activation by microinjection of sperm factor
1- Inject oocytes with 10 to 12 picoliters of sperm factor
isolated, e.g., from primates, pigs, bovine, sheep, goats,
horses, mice, rats, rabbits or hamsters;
2- Put eggs in culture.
8. Activation by microinjection of recombinant sperm factor.
9. Activation by Exposure to DMAP followed by Cycloheximide and Cytochalasin B
Place oocytes or NT units, typically about 22 to 28 hours post
maturation in about 2 mM DMAP for about one hour, followed by
incubation for about two to twelve hours, preferably about eight
hours, in 5 μg/ml of cytochalasin B and 20 μg/ml cycloheximide. The above activation protocols are exemplary of protocols used for nuclear
transfer procedures, e.g., those including the use of primate or human donor cells
or oocytes. However, the above activation protocols may be used when either or
both the donor cell and nucleus is of ungulate origin, e.g., a sheep, buffalo, horse,
goat, bovine, pig and or wherein the oocyte is of ungulate origin, e.g., sheet, pig,
buffalo, horse, goat, bovine, etc., as well as for other species.
As noted, activation may be effected before, simultaneous, or after nuclear
transfer. In general, activation will be effected about 40 hours prior to nuclear
transfer and fusion to about 40 hours after nuclear transfer and fusion, more
preferably about 24 hours before to about 24 hours after nuclear transfer and
fusion, and most preferably from about 4 to 9 hours before nuclear transfer and
fusion to about 4 to 9 hours after nuclear transfer and fusion. Activation is
preferably effected after or proximate to in vitro or in vivo maturation of the
oocyte, e.g., approximately simultaneous or within about 40 hours of maturation,
more preferably within about 24 hours of maturation.
Activated NT units may be cultured in a suitable in vitro culture medium
until the generation of embryonic or stem-like cells and cell colonies. Culture
media suitable for culturing and maturation of embryos are well known in the art.
Examples of known media, which may be used for bovine embryo culture and
maintenance, include Ham's F-10 + 10% fetal calf serum (FCS), Tissue Culture
Medium- 199 (TCM-199) + 10% fetal calf serum, Tyrodes-Albumin-Lactate- Pyruvate (TALP), Dulbecco's Phosphate Buffered Saline (PBS), Eagle's and
Whitten's media. One of the most common media used for the collection and
maturation of oocytes is TCM-199, and 1 to 20% serum supplement including fetal
calf serum, newborn serum, estrual cow serum, lamb serum or steer serum. A
preferred maintenance medium includes TCM-199 with Earl salts, 10% fetal calf
serum, 0.2 Ma pyruvate and 50 μg/ml gentamicin sulphate. Any of the above may
also involve co-culture with a variety of cell types such as granulosa cells, oviduct
cells, BRL cells and uterine cells and STO cells.
In particular, human epithelial cells of the endometrium secrete leukemia
inhibitory factor (LIF) during the preimplantation and implantation period.
Therefore, the addition of LIF to the culture medium could be of importance in
enhancing the in vitro development of the reconstructed embryos. The use of LIF
for embryonic or stem-like cell cultures has been described in U.S. Patent
5,712,156, which is herein incorporated by reference.
Another maintenance medium is described in U.S. Patent 5,096,822 to
Rosenkrans, Jr. et al., which is incorporated herein by reference. This embryo
medium, named CR1, contains the nutritional substances necessary to support an
embryo. CR1 contains hemicalcium L-lactate in amounts ranging from 1.0 mM
to 10 mM, preferably 1.0 mM to 5.0 mM. Hemicalcium L-lactate is L-lactate with
a hemicalcium salt incorporated thereon. Also, suitable culture medium for maintaining human embryonic cells in
culture as discussed in Thomson et al., Science, 282: 1145-1147 (1998) and Proc.
Natl Acad. Set, USA, 92:7844-7848 (1995).
Afterward, the cultured NT unit or units are preferably washed and then
placed in a suitable media, e.g., CRIaa medium, Ham's F-10, Tissue Culture Media
-199 (TCM-199). Tyrodes-Albumin-Lactate-Pyruvate (TALP) Dulbecco's
Phosphate Buffered Saline (PBS), Eagle's or Whitten's, preferably containing
about 10% FCS. Such culturing will preferably be effected in well plates which
contain a suitable confluent feeder layer. Suitable feeder layers include, by way
of example, fibroblasts and epithelial cells, e.g., fibroblasts and uterine epithelial
cells derived from ungulates, chicken fibroblasts, murine (e.g., mouse or rat) fibro¬
blasts, STO and SI-m220 feeder cell lines, and BRL cells.
In the preferred embodiment, the feeder cells will comprise mouse
embryonic fibroblasts. Means for preparation of a suitable fibroblast feeder layer
are described in the example which follows and is well within the skill of the
ordinary artisan.
The NT units are cultured on the feeder layer until the NT units reach a size
suitable for obtaining cells which may be used to produce embryonic stem-like
cells or cell colonies. Preferably, these NT units will be cultured until they reach
a size of at least about 2 to 400 cells, more preferably about 4 to 128 cells, and
most preferably at least about 50 cells. The culturing will be effected under suit¬
es - able conditions, i.e., about 38.5°C and 5% C02, with the culture medium changed
in order to optimize growth typically about every 2-5 days, preferably about every
3 days.
In the case of human cell/enucleated bovine oocyte derived NT units,
sufficient cells to produce an ES cell colony, typically on the order of about 50
cells, will be obtained about 12 days after initiation of oocyte activation.
However, this may vary dependent upon the particular cell used as the nuclear
donor, the species of the particular oocyte, and culturing conditions. One skilled
in the art can readily ascertain visually when a desired sufficient number of cells
has been obtained based on the morphology of the cultured NT units.
In the case of human/human nuclear transfer embryos, or other embryos
produced using non-human primate donor or oocyte, it may be advantageous to use
culture medium known to be useful for maintaining human and other primate cells
in tissue culture. Examples of a culture media suitable for human embryo culture
include the medium reported in Jones et al, Human Reprod., 13(1):169-177 (1998),
the PI -catalog #99242 medium, and the P-l catalog #99292 medium, both
available from Irvine Scientific, Santa Ana, California, and those used by
Thomson et al. (1998) and (1995), which references are incorporated by reference
in their entirety.
Another preferred medium comprises ACM + uridine + glucose + 1000 IU
of LIF. As discussed above, the cells used in the present invention will preferably comprise mammalian somatic cells, most preferably cells derived from
an actively proliferating (non-quiescent) mammalian cell culture. In an especially
preferred embodiment, the donor cell will be genetically modified by the addition,
deletion or substitution of a desired DNA sequence. For example, the donor cell,
e.g., a keratinocyte or fibroblast, e.g., of human, primate or bovine origin, may be
transfected or transformed with a DNA construct that provides for the expression
of a desired gene product, e.g., therapeutic polypeptide. Examples thereof include
lymphokines, e.g., IGF-I, IGF-II, interferons, colony stimulating factors,
connective tissue polypeptides such as collagens, genetic factors, clotting factors,
enzymes, enzyme inhibitors, etc.
Also, as discussed above, the donor cells may be modified prior to nuclear
transfer, e.g., to effect impaired cell lineage development, enhanced embryonic
development and or inhibition of apoptosis. Examples of desirable modifications
are discussed further below.
One aspect of the invention will involve genetic modification of the donor
cell, e.g., a human cell, such that it is lineage deficient and therefore when used for
nuclear transfer it will be unable to give rise to a viable offspring. This is
desirable especially in the context of human nuclear transfer embryos, wherein for
ethical reasons, production of a viable embryo may be an unwanted outcome. This
can be effected by genetically engineering a human cell such that it is incapable
of differentiating into specific cell lineages when used for nuclear transfer. In particular, cells may be genetically modified such that when used as nuclear
transfer donors the resultant "embryos" do not contain or substantially lack at least
one of mesoderm, endoderm or ectoderm tissue.
This can be accomplished by, e.g., knocking out or impairing the
expression of one or more mesoderm, endoderm or ectoderm specific genes.
Examples thereof include:
Mesoderm: SRF, MESP-1 , HNF-4, beta-I integrin, MSD;
Endoderm: GATA-6, GATA-4;
Ectoderm: RNA helicase A, H beta 58.
The above list is intended to be exemplary and non-exhaustive of known
genes which are involved in the development of mesoderm, endoderm and
ectoderm. The generation of mesoderm deficient, endoderm deficient and
ectoderm deficient cells and embryos has been previously reported in the literature.
See, e.g., Arsenian et al, EMBO J., Vol. 17(2):6289-6299 (1998); Saga Y, Mech.
Dev., Vol. 75(l-2):53-66 (1998); Holdener et al, Development, Vol. 120(5): 1355-
1346 (1994); Chen et al, Genes Dev. Vol. 8(20):2466-2477 (1994); Rohwedel et
al, Dev. Biol, 201(2):167-189 (1998) (mesoderm); Morrisey et al, Genes, Dev.,
Vol. 12(22):3579-3590 (1998); Soudais et al, Development, Vol. 121(11):3877-
3888 (1995) (endoderm); and Lee et al, Proc. Natl. Acad. Set USA, Vol.
95:(23):13709-13713 (1998); andRadice et al, Development, Vol. 111(3):801-811
(1991) (ectoderm). In general, a desired somatic cell, e.g., a human keratinocyte, epithelial cell
or fibroblast, will be genetically engineered such that one or more genes specific
to particular cell lineages are "knocked out" and/or the expression of such genes
significantly impaired. This may be effected by known methods, e.g., homologous
recombination. A preferred genetic system for effecting "knock-out" of desired
genes is disclosed by Capecchi et al, U.S. Patents 5,631,153 and 5,464,764, which
reports positive-negative selection (PNS) vectors that enable targeted modification
of DNA sequences in a desired mammalian genome. Such genetic modification
will result in a cell that is incapable of differentiating into a particular cell lineage
when used as a nuclear transfer donor.
This genetically modified cell will be used to produce a lineage-defective
nuclear transfer embryo, i.e., that does not develop at least one of a functional
mesoderm, endoderm or ectoderm. Thereby, the resultant embryos, even if
implanted, e.g., into a human uterus, would not give rise to a viable offspring.
However, the ES cells that result from such nuclear transfer will still be useful in
that they will produce cells of the one or two remaining non-impaired lineage. For
example, an ectoderm deficient human nuclear transfer embryo will still give rise
to mesoderm and endoderm derived differentiated cells. An ectoderm deficient
cell can be produced by deletion and/or impairment of one or both of RNA
helicase A or H beta 58 genes. These lineage deficient donor cells may also be genetically modified to
express another desired DNA sequence.
Thus, the genetically modified donor cell will give rise to a lineage-
deficient blastocyst which, when plated, will differentiate into at most two of the
embryonic germ layers.
Alternatively, the donor cell can be modified such that it is "mortal". This
can be achieved by expressing anti-sense or ribozyme telomerase genes. This can
be effected by known genetic methods that will provide for expression of antisense
DNA or ribozymes, or by gene knockout. These "mortal" cells, when used for
nuclear transfer, will not be capable of differentiating into viable offspring.
Another preferred embodiment of the present invention is the production
of nuclear transfer embryos that grow more efficiently in tissue culture. This is
advantageous in that it should reduce the requisite time and necessary fusions to
produce ES cells and/or offspring (if the blastocysts are to be implanted into a
female surrogate). This is desirable also because it has been observed that
blastocysts and ES cells resulting from nuclear transfer may have impaired
development potential. While these problems may often be alleviated by alteration
of tissue culture conditions, an alternative solution is to enhance embryonic
development by enhancing expression of genes involved in embryonic
development. For example, it has been reported that the gene products of the Ped type,
which are members of the MHC I family, are of significant importance to
embryonic development. More specifically, it has been reported in the case of
mouse preimplantation embryos that the Q7 and Q9 genes are responsible for the
"fast growth" phenotype. Therefore, it is anticipated that introduction of DNAs
that provide for the expression of these and related genes, or their human or other
mammalian counterparts into donor cells, will give rise to nuclear transfer embryos
that grow more quickly. This is particularly desirable in the context of cross-
species nuclear transfer embryos which may develop less efficiently in tissue
culture than nuclear transfer embryos produced by fusion of cells or nuclei of the
same species.
In particular, a DNA construct containing the Q7 and/or Q9 gene will be
introduced into donor somatic cells prior to nuclear transfer. For example, an
expression construct can be constructed containing a strong constitutive
mammalian promoter operably linked to the Q7 and/or Q9 genes, an IRES, one or
more suitable selectable markers, e.g,. neomycin, ADA, DHFR, and a poly-A
sequence, e.g., bGH polyA sequence. Also, it may be advantageous to further
enhance Q7 and Q9 gene expression by the inclusion of insulates. It is anticipated
that these genes will be expressed early on in blastocyst development as these
genes are highly conserved in different species, e.g., bovines, goats, porcine, dogs,
cats, and humans. Also, it is anticipated that donor cells can be engineered to affect other genes that enhance embryonic development. Thus, these genetically
modified donor cells should produce blastocysts and preimplantation stage
embryos more efficiently.
Still another aspect of the invention involves the construction of donor cells
that are resistant to apoptosis, i.e., programmed cell death. It has been reported in
the literature that cell death related genes are present in preimplantation stage
embryos. (Adams et al, Science, 281(5381):1322-1326 (1998)). Genes reported
to induce apoptosis include, e.g., Bad, Bok, BH3, Bik, Hrk, BNIP3, BimL, Bad,
Bid, and EGL-1. By contrast, genes that reportedly protect cells from programmed
cell death include, by way of example, BcL-XL, Bcl-w, Mcl-l, Al, Nr-13, BHRF-
1, LMW5-HL, ORF16, Ks-Bel-2, E1B-19K, and CED-9.
Thus, donor cells can be constructed wherein genes that induce apoptosis
are "knocked out" or wherein the expression of genes that protect the cells from
apoptosis is enhanced or turned on during embryonic development.
For example, this can be effected by introducing a DNA construct that
provides for regulated expression of such protective genes, e.g., Bcl-2 or related
genes during embryonic development. Thereby, the gene can be "turned on" by
culturing the embryo under specific growth conditions. Alternatively, it can be
linked to a constitutive promoter.
More specifically, a DNA construct containing a Bcl-2 gene operably
linked to a regulatable or constitutive promoter, e.g., PGK, SV40, CMV, ubiquitin, or beta-actin, an IRES, a suitable selectable marker, and a poly-A sequence can be
constructed and introduced into a desired donor mammalian cell, e.g., human
keratinocyte or fibroblast.
These donor cells, when used to produce nuclear transfer embryos, should
be resistant to apoptosis and thereby differentiate more efficiently in tissue culture.
Thereby, the speed and/or number of suitable preimplantation embryos produced
by nuclear transfer can be increased.
Another means of accomplishing the same result is to impair the expression
of one or more genes that induce apoptosis. This will be effected by knock-out or
by the use of antisense or ribozymes against genes that are expressed in and which
induce apoptosis early on in embryonic development. Examples thereof are
identified above. Cell death genes that may be expressed in the antisense
orientation include BAX, Apaf-1, and capsases. Additionally, a transgene may be
introduced that encodes for methylase or demethylase in the sense or antisense
orientation. DNAs that encode methylase and demethylase enzymes are well
known in the art. Still alternatively, donor cells may be constructed containing
both modifications, i.e., impairment of apoptosis-inducing genes and enhanced
expression of genes that impede or prevent apoptosis. The construction and
selection of genes that affect apoptosis, and cell lines that express such genes, is
disclosed in U.S. Patent No. 5,646,008, which patent is incorporated by reference herein. Many DNAs that promote or inhibit apoptosis have been reported and are
the subject of numerous patents.
Another means of enhancing cloning efficiency is to select cells of a
particular cell cycle stage as the donor cell. It has been reported that this can have
significant effects on nuclear transfer efficiency. (Barnes et al, Mol. Reprod.
Devel, 36(1):33-41 (1993). Different methods for selecting cells of a particular
cell cycle stage have been reported and include serum starvation (Campbell et al,
Nature, 380:64-66 (1996); Wilmut et al, Nature, 385:810-813 (1997), and
chemical synchronization (Urbani et al, Exp. Cell Res., 219(1):159-168 (1995).
For example, a particular cyclin DNA may be operably linked to a regulatory
sequence, together with a detectable marker, e.g., green fluorescent protein (GFP),
followed by the cyclin destruction box, and optionally insulation sequences to
enhance cyclin and marker protein expression. Thereby, cells of a desired cell
cycle can be easily visually detected and selected for use as a nuclear transfer
donor. An example thereof is the cyclin Dl gene in order to select for cells that
are in Gl. However, any cyclin gene should be suitable for use in the claimed
invention. (See, e.g., King et al, Mol. Biol. Cell, Vol. 7(9): 1343-1357 (1996)).
However, a less invasive or more efficient method for producing cells of
a desired cell cycle stage are needed. It is anticipated that this can be effected by
genetically modifying donor cells such that they express specific cyclins under detectable conditions. Thereby, cells of a specific cell cycle can be readily
discerned from other cell cycles.
Cyclins are proteins that are expressed only during specific stages of the
cell cycle. They include cyclin Dl, D2 and D3 in Gl phase, cyclin BI and B2 in
G2/M phase and cyclin E, A and H in S phase. These proteins are easily translated
and destroyed in the cytogolcytosol. This "transient" expression of such proteins
is attributable in part to the presence of a "destruction box", which is a short amino
acid sequence that is part of the protein that functions as a tag to direct the prompt
destruction of these proteins via the ubiquitin pathway. (Adams et al, Science, 281
(5321):1322-1326 (1998)).
In the present invention, donor cells will be constructed that express one or
more of such cyclin genes under easily detectable conditions, preferably
visualizable, e.g., by the use of a fluorescent label. For example, a particular
cyclin DNA may be operably linked to a regulatory sequence, together with a
detectable marker, e.g., green fluorescent protein (GFP), followed by the cyclin
destruction box, and optionally insulation sequences to enhance cyclin and/or
marker protein expression. Thereby, cells of a desired cell cycle can be easily
visually detected and selected for use as a nuclear transfer donor. An example
thereof is the cyclin Dl gene which can be used to select for cells that are in Gl.
However, any cyclin gene should be suitable for use in the claimed invention.
(See, e.g., King et al, Mol. Biol. Cell, Vol. 7(9):1343-1357 (1996)). As discussed, the present invention provides different methods for
enhancing nuclear transfer efficiency, preferably a cross-species nuclear transfer
process. While the present inventors have demonstrated that nuclei or cells of one
species when inserted or fused with an enucleated oocyte of a different species can
give rise to nuclear transfer embryos that produce blastocysts, which embryos can
give rise to ES cell lines, the efficiency of such process is quite low. Therefore,
many fusions typically need to be effected to produce a blastocyst the cells of
which may be cultured to produce ES cells and ES cell lines. Yet another means
for enhancing the development of nuclear transfer embryos in vitro is by
optimizing culture conditions. One means of achieving this result will be to
culture NT embryos under conditions impede apoptosis. With respect to this
embodiment of the invention, it has been found that proteases such as capsases can
cause oocyte death by apoptosis similar to other cell types. (See, Jurisicosva et al,
Mol. Reprod. Devel, 51(3):243-253 (1998).)
It is anticipated that blastocyst development will be enhanced by including
in culture media used for nuclear transfer and to maintain blastocysts or culture
preimplantation stage embryos one or more capsase inhibitors. Such inhibitors
include by way of example capsase-4 inhibitor I, capsase-3 inhibitor I, capsase-6
inhibitor II, capsase-9 inhibitor II, and capsase- 1 inhibitor I. The amount thereof
will be an amount effective to inhibit apoptosis, e.g., 0.00001 to 5.0% by weight
of medium; more preferably 0.01%> to 1.0% by weight of medium. Thus, the foregoing methods may be used to increase the efficiency of nuclear transfer by
enhancing subsequent blastocyst and embryo development in tissue culture.
After NT units of the desired size are obtained, the cells are mechanically
removed from the zone and are then used to produce embryonic or stem-like cells
and cell lines. This is preferably effected by taking the clump of cells which
comprise the NT unit, which typically will contain at least about 50 cells, washing
such cells, and plating the cells onto a feeder layer, e.g., irradiated fibroblast cells.
Typically, the cells used to obtain the stem-like cells or cell colonies will be
obtained from the inner most portion of the cultured NT unit which is preferably
at least 50 cells in size. However, NT units of smaller or greater cell numbers as
well as cells from other portions of the NT unit may also be used to obtain ES-like
cells and cell colonies.
It is further envisioned that a longer exposure of donor cell DNA to the
oocyte's cytosol may facilitate the dedifferentiation process. This can be
accomplished by re-cloning, i.e., by taking blastomeres from a reconstructed
embryo and fusing them with a new enucleated oocyte. Alternatively, the donor
cell may be fused with an enucleated oocyte and four to six hours later, without
activation, chromosomes removed and fused with a younger oocyte. Activation
would occur thereafter.
The cells are maintained in the feeder layer in a suitable growth medium,
e.g., alpha MEM supplemented with 10% FCS and 0.1 mM beta-mercaptoethanol (Sigma) and L-glutamine. The growth medium is changed as often as necessary
to optimize growth, e.g., about every 2-3 days.
This culturing process results in the formation of embryonic or stem-like
cells or cell lines. In the case of human cell/bovine oocyte derived NT embryos,
colonies are observed by about the second day of culturing in the alpha MEM
medium. However, this time may vary dependent upon the particular nuclear
donor cell, specific oocyte and culturing conditions. One skilled in the art can vary
the culturing conditions as desired to optimize growth of the particular embryonic
or stem-like cells. Other suitable media are disclosed herein.
The embryonic or stem-like cells and cell colonies obtained will typically
exhibit an appearance similar to embryonic or stem-like cells of the species used
as the nuclear cell donor rather than the species of the donor oocyte. For example,
in the case of embryonic or stem-like cells obtained by the transfer of a human
nuclear donor cell into an enucleated bovine oocyte, the cells exhibit a morphology
more similar to mouse embryonic stem cells than bovine ES-like cells.
More specifically, the individual cells of the human ES-line cell colony are
not well defined, and the perimeter of the colony is refractive and smooth in
appearance. Further, the cell colony has a longer cell doubling time, about twice
that of mouse ES cells. Also, unlike bovine and porcine derived ES cells, the
colony does not possess an epithelial-like appearance. As discussed above, it has been reported by Thomson, in U.S. Patent
5,843,780, that primate stem cells are SSEA-1 (-), SSEA-4 (+), TRA-1-60 (+),
TRA-1-81 (+) and alkaline phosphatase (+). It is anticipated that human and
primate ES cells produced according to the present methods will exhibit similar
or identical marker expression.
Alternatively, that such cells are actual human or primate embryonic stem
cells will be confirmed based on their capability of giving rise to all of mesoderm,
ectoderm and endoderm tissues. This will be demonstrated by culturing ES cells
produced according to the invention under appropriate conditions, e.g., as
disclosed by Thomsen, U.S. Patent 5,843,780, incorporated by reference in its
entirety herein. Alternatively, the fact that the cells produced according to the
invention are pluripotent will be confirmed by injecting such cells into an animal,
e.g., a SCID mouse, or large agricultural animal, and thereafter obtaining tissues
that result from said implanted cells. These implanted ES cells should give rise
to all different types of differentiated tissues, i.e., mesoderm, ectoderm, and
endodermal tissues.
The resultant embryonic or stem-like cells and cell lines, preferably human
embryonic or stem-like cells and cell lines, have numerous therapeutic and
diagnostic applications. Most especially, such embryonic or stem-like cells may
be used for cell transplantation therapies. Human embryonic or stem-like cells
have application in the treatment of numerous disease conditions. Still another object of the present invention is to improve the efficacy of
nuclear transfer, e.g., cross-species nuclear transfer by introducing mitochondrial
DNA of the same species as the donor cell or nucleus into the recipient oocyte
before or after nuclear transfer, before or after activation, and before or after
5 fusion and cleavage. Preferably, if the donor cell is human, human mitochondrial
DNA will be derived from cells of the particular donor, e.g., liver cells and tissue.
Methods for isolating mitochondria are well known in the art.
Mitochondria can be isolated from cells in tissue culture, or from tissue. The
particular cells or tissue will depend upon the particular species of the donor cell.
o Examples of cells or tissues that may be used as sources of mitochondria include
fibroblasts, epithelium, liver, lung, keratinocyte, stomach, heart, bladder, pancreas,
esophageal, lymphocytes, monocytes, mononuclear cells, cumulus cells, uterine
cells, placental cells, intestinal cells, hematopoietic cells, and tissues containing
such cells.
5 For example, mitochondria can be isolated from tissue culture cells and rat
liver. It is anticipated that the same or similar procedures may be used to isolate
mitochondria from other cells and tissues. As noted above, preferred source of
mitochondria comprises human liver tissue because such cells contain a large
number of mitochondria. Those skilled in the art will be able to modify the
o procedure as necessary, dependent upon the particular cell line or tissue. The isolated DNA can also be further purified, if desired, known methods, e.g., density
gradient centrifugation.
In this regard, it is known that mouse embryonic stem (ES) cells are
capable of differentiating into almost any cell type, e.g., hematopoietic stem cells.
Therefore, human embryonic or stem-like cells produced according to the
invention should possess similar differentiation capacity. The embryonic or stem¬
like cells according to the invention will be induced to differentiate to obtain the
desired cell types according to known methods. For example, the subject human
embryonic or stem-like cells may be induced to differentiate into hematopoietic
stem cells, muscle cells, cardiac muscle cells, liver cells, cartilage cells, epithelial
cells, urinary tract cells, etc., by culturing such cells in differentiation medium and
under conditions which provide for cell differentiation. Medium and methods
which result in the differentiation of embryonic stem cells are known in the art as
are suitable culturing conditions.
For example, Palacios et al, Proc. Natl. Acad. Set, USA, 92:7530-7537
(1995) teaches the production of hematopoietic stem cells from an embryonic cell
line by subjecting stem cells to an induction procedure comprising initially
culturing aggregates of such cells in a suspension culture medium lacking retinoic
acid followed by culturing in the same medium containing retinoic acid, followed
by transferral of cell aggregates to a substrate which provides for cell attachment. Moreover, Pedersen, J. Reprod. Fertil. Dev., 6:543-552 (1994) is a review
article which references numerous articles disclosing methods for in vitro
differentiation of embryonic stem cells to produce various differentiated cell types
including hematopoietic cells, muscle, cardiac muscle, nerve cells, among others.
Further, Bain et al, Dev. Biol, 168:342-357 (1995) teaches in vitro
differentiation of embryonic stem cells to produce neural cells which possess
neuronal properties. These references are exemplary of reported methods for
obtaining differentiated cells from embryonic or stem-like cells. These references
and in particular the disclosures therein relating to methods for differentiating
embryonic stem cells are incorporated by reference in their entirety herein.
Thus, using known methods and culture medium, one skilled in the art may
culture the subject embryonic or stem-like cells to obtain desired differentiated cell
types, e.g., neural cells, muscle cells, hematopoietic cells, etc. In addition, the use
of inducible Bcl-2 or Bcl-xl might be useful for enhancing in vitro development
of specific cell lineages. In vivo, Bcl-2 prevents many, but not all, forms of
apoptotic cell death that occur during lymphoid and neural development. A
thorough discussion of how Bcl-2 expression might be used to inhibit apoptosis
of relevant cell lineages following transfection of donor cells is disclosed in U.S.
Patent No. 5,646,008, which is herein incorporated by reference.
The subject embryonic or stem-like cells may be used to obtain any desired
differentiated cell type. Therapeutic usages of such differentiated human cells are unparalleled. For example, human hematopoietic stem cells may be used in
medical treatments requiring bone marrow transplantation. Such procedures are
used to treat many diseases, e.g., late stage cancers such as ovarian cancer and
leukemia, as well as diseases that compromise the immune system, such as AIDS.
Hematopoietic stem cells can be obtained, e.g., by fusing adult somatic cells of
a cancer or AIDS patient, e.g., epithelial cells or lymphocytes with an enucleated
oocyte, e.g., bovine oocyte, obtaining embryonic or stem-like cells as described
above, and culturing such cells under conditions which favor differentiation, until
hematopoietic stem cells are obtained. Such hematopoietic cells may be used in
the treatment of diseases including cancer and AIDS.
Alternatively, adult somatic cells from a patient with a neurological
disorder may be fused with an enucleated animal oocyte, e.g., a primate or bovine
oocyte, human embryonic or stem-like cells obtained therefrom, and such cells
cultured under differentiation conditions to produce neural cell lines. Specific dis-
eases treatable by transplantation of such human neural cells include, by way of
example, Parkinson's disease, Alzheimer's disease, ALS and cerebral palsy, among
others. In the specific case of Parkinson's disease, it has been demonstrated that
transplanted fetal brain neural cells make the proper connections with surrounding
cells and produce dopamine. This can result in long-term reversal of Parkinson's
disease symptoms. To allow for specific selection of differentiated cells, donor cells may be
transfected with selectable markers expressed via inducible promoters, thereby
permitting selection or enrichment of particular cell lineages when differentiation
is induced. For example, CD34-neo may be used for selection of hematopoietic
cells, Pwl-neo for muscle cells, Mash-1-neo for sympathetic neurons, Mal-neo for
human CNS neurons of the grey matter of the cerebral cortex, etc.
The great advantage of the subject invention is that it provides an
essentially limitless supply of isogenic or synegenic human cells suitable for trans¬
plantation. Therefore, it will obviate the significant problem associated with
current transplantation methods, i.e., rejection of the transplanted tissue which may
occur because of host-vs-graft or graft- vs-host rejection. Conventionally, rejection
is prevented or reduced by the administration of anti-rejection drugs such as
cyclosporin. However, such drugs have significant adverse side-effects, e.g.,
immunosuppression, carcinogenic properties, as well as being very expensive.
The present invention should eliminate, or at least greatly reduce, the need for anti-
rejection drugs, such as cyclosporine, imulan, FK-506, glucocorticoids, and
rapamycin, and derivatives thereof.
Other diseases and conditions treatable by isogenic cell therapy include, by
way of example, spinal cord injuries, multiple sclerosis, muscular dystrophy,
diabetes, liver diseases, i.e., hypercholesterolemia, heart diseases, cartilage
replacement, burns, foot ulcers, gastrointestinal diseases, vascular diseases, kidney
disease, urinary tract disease, and aging related diseases and conditions.
Also, human embryonic or stem-like cells produced according to the
invention may be used to produce genetically engineered or transgenic human
differentiated cells. Essentially, this will be effected by introducing a desired gene
or genes, which may be heterologous, or removing all or part of an endogenous
gene or genes of human embryonic or stem-like cells produced according to the
invention, and allowing such cells to differentiate into the desired cell type. A
preferred method for achieving such modification is by homologous recombination
because such technique can be used to insert, delete or modify a gene or genes at
a specific site or sites in the stem-like cell genome.
This methodology can be used to replace defective genes, e.g., defective
immune system genes, cystic fibrosis genes, or to introduce genes which result in
the expression of therapeutically beneficial proteins such as growth factors,
lymphokines, cytokines, enzymes, etc. For example, the gene encoding brain
derived growth factor may be introduced into human embryonic or stem-like cells,
the cells differentiated into neural cells and the cells transplanted into a
Parkinson's patient to retard the loss of neural cells during such disease. Previously, cell types transfected with BDNF varied from primary cells to
immortalized cell lines, either neural or non-neural (myoblast and fibroblast)
derived cells. For example, astrocytes have been transfected with BDNF gene
using retroviral vectors, and the cells grafted into a rat model of Parkinson's
disease (Yoshimoto et al., Brain Research, 691:25-36, (1995)).
This ex vivo therapy reduced Parkinson's-like symptoms in the rats up to
45% 32 days after transfer. Also, the tyrosine hydroxylase gene has been placed
into astrocytes with similar results (Lundberg et al., Develop. Neurol, 139:39-53
(1996) and references cited therein).
However, such ex vivo systems have problems. In particular, retroviral
vectors currently used are down-regulated in vivo and the transgene is only
transiently expressed (review by Mulligan, Science, 260:926-932 (1993)). Also,
such studies used primary cells, astrocytes, which have finite life span and
replicate slowly. Such properties adversely affect the rate of transfection and
impede selection of stably transfected cells. Moreover, it is almost impossible to
propagate a large population of gene targeted primary cells to be used in homolo¬
gous recombination techniques.
By contrast, the difficulties associated with retroviral systems should be
eliminated by the use of human embryonic or stem-like cells. It has been demon-
strated previously by the subject assignee that cattle and pig embryonic cell lines
can be transfected and selected for stable integration of heterologous DNA. Such methods are described in commonly assigned U.S. Serial No. 08/626,054, filed
April 1, 1996, now U.S. Patent No. 5,905,042, incorporated by reference in its
entirety. Therefore, using such methods or other known methods, desired genes
may be introduced into the subject human embryonic or stem-like cells, and the
cells differentiated into desired cell types, e.g., hematopoietic cells, neural cells,
pancreatic cells, cartilage cells, etc.
Genes which may be introduced into the subject embryonic or stem-like
cells include, by way of example, epidermal growth factor, basic fibroblast growth
factor, glial derived neurotrophic growth factor, insulin-like growth factor (I and
II), neurotiophin-3, neurotiOphin-4/5, ciliary neurotrophic factor, AFT- 1 , cytokine
genes (interleukins, interferons, colony stimulating factors, tumor necrosis factors
(alpha and beta), etc.), genes encoding therapeutic enzymes, collagen, human
serum albumin, etc.
In addition, it is also possible to use one of the negative selection systems
now known in the art for eliminating therapeutic cells from a patient if necessary.
For example, donor cells transfected with the thymidine kinase (TK) gene will
lead to the production of embryonic cells containing the TK gene. Differentiation
of these cells will lead to the isolation of therapeutic cells of interest which also
express the TK gene. Such cells may be selectively eliminated at any time from
a patient upon gancyclovir administration. Such a negative selection system is
described in U.S. Patent No. 5,698,446, and is herein incorporated by reference. The subject embryonic or stem-like cells, preferably human cells, also may
be used as an in vitro model of differentiation, in particular for the study of genes
which are involved in the regulation of early development.
Also, differentiated cell tissues and organs using the subject embryonic or
stem-like cells may be used in drug studies.
Further, the subject cells may be used to express recombinant DNAs.
Still further, the subject embryonic or stem-like cells may be used as
nuclear donors for the production of other embryonic or stem-like cells and cell
colonies.
Also, cultured inner cell mass, or stem cells, produced according to the
invention may be introduced into animals, e.g., SCID mice, cows, pigs, e.g., under
the renal capsule or intramuscularly and used to produce a teratoma therein. This
teratoma can be used to derive different tissue types. Also, the inner cell mass
produced by X-species nuclear transfer may be introduced together with a
biodegradable, biocompatible polymer matrix that provides for the formation of
3 -dimensional tissues. After tissue formation, the polymer degrades, ideally just
leaving the donor tissue, e.g., cardiac, pancreatic, neural, lung, liver. In some
instances, it may be advantageous to include growth factors and proteins that
promote angiogenesis. Alternatively, the formation of tissues can be effected
totally in vitro, with appropriate culture media and conditions, growth factors, and
biodegradable polymer matrices. In order to more clearly describe the subject invention, the following
examples are provided.
EXAMPLE 1
MATERIALS AND METHODS
Donor Cells for Nuclear Transfer
Epithelial cells were lightly scraped from the inside of the mouth of a
consenting adult with a standard glass slide. The cells were washed off the slide
into a petri dish containing phosphate buffered saline without Ca or Mg. The cells
were pipetted through a small-bore pipette to break up cell clumps into a single
cell suspension. The cells were then transferred into a microdrop of TL-HEPES
medium containing 10% fetal calf serum (FCS) under oil for nuclear transfer into
enucleated cattle oocytes.
Nuclear Transfer Procedures
Basic nuclear transfer procedures have been described previously. Briefly,
after slaughterhouse oocytes were matured in vitro the oocytes were stripped of
cumulus cells and enucleated with a beveled micropipette at approximately 18
hours post maturation (hpm). Enucleation was confirmed in TL-HEPES medium
plus bisbenzimide (Hoechst 33342, 3 μg/ml; Sigma). Individual donor cells were
then placed into the perivitelline space of the recipient oocyte. The bovine oocyte
cytoplasm and the donor nucleus (NT unit) are fused together using electrofusion
techniques. One fusion pulse consisting of 90 V for 15 μsec was applied to the NT
unit. This occurred at 24 hours post-initiation of maturation (hpm) of the oocytes.
The NT units were placed in CR1 aa medium until 28 hpm.
The procedure used to artificially activate oocytes has been described
elsewhere. NT unit activation was at 28 hpm. A brief description of the activation
procedure is as follows: NT units were exposed for four min to ionomycin (5 μM;
CalBiochem, La Jolla, CA) in TL-HEPES supplemented with 1 mg/ml BSA and
then washed for five min in TL-HEPES supplemented with 30 mg/ml BSA. The
NT units were then transferred into a microdrop of CRlaa culture medium
containing 0.2 mM DMAP (Sigma) and cultured at 38.5°C 5% C02 for four to five
hours. The NT units were washed and then placed in a CRlaa medium plus 10%
FCS and 6 mg/ml BSA in four well plates containing a confluent feeder layer of
mouse embryonic fibroblasts (described below). The NT units were cultured for
three more days at 38.5°C and 5% C02. The culture medium was changed every three days until day 12 after the time of activation. At this time NT units reaching
the desired cell number, i.e., about 50 cell number, were mechanically removed
from the zona and used to produce embryonic cell lines. A photograph of an NT
unit obtained as described above is contained in Figure 1.
Fibroblast feeder layer
Primary cultures of embryonic fibroblasts were obtained from 14-16 day
old murine fetuses. After the head, liver, heart and alimentary tract were
aseptically removed, the embryos were minced and incubated for 30 minutes at
37°C in pre-warmed trypsin EDTA solution (0.05% trypsin 0.02% EDTA;
GIBCO, Grand Island, NY). Fibroblast cells were plated in tissue culture flasks
and cultured in alpha-MEM medium (BioWhittaker, Walkersville, MD) supple¬
mented with 10% fetal calf serum (FCS) (Hyclone, Logen, UT), penicillin (100
IU/ml) and streptomycin (50 μl/ml). Three to four days after passage, embryonic
fibroblasts, in 35 x 10 Nunc culture dishes (Baxter Scientific, McGaw Park, IL),
were irradiated. The irradiated fibroblasts were grown and maintained in a
humidified atmosphere with 5% C02 in air at 37°C. The culture plates which had
a uniform monolayer of cells were then used to culture embryonic cell lines.
Production of embryonic cell line.
NT unit cells obtained as described above were washed and plated directly
onto irradiated feeder fibroblast cells. These cells included those of the inner
portion of the NT unit. The cells were maintained in a growth medium consisting
of alpha MEM supplemented with 10% FCS and 0.1 mM beta-mercaptoethanol
(Sigma). Growth medium was exchanged every two to three days. The initial
colony was observed by the second day of culture. The colony was propagated
and exhibits a similar morphology to previously disclosed mouse embryonic stem
(ES) cells. Individual cells within the colony are not well defined and the
perimeter of the colony is refractile and smooth in appearance. The cell colony
appears to have a slower cell doubling time than mouse ES cells. Also, unlike
bovine and porcine derived ES cells, the colony does not have an epithelial
appearance thus far. Figures 2 through 5 are photographs of ES-like cell colonies
obtained as described, supra.
Production of Differentiated Human Cells
The human embryonic cells obtained are transferred to a differentiation
medium and cultured until differentiated human cell types are obtained.
RESULTS
Table 1. Human cells as donor nuclei in NT unit production and development.
I TABLE 1
Figure imgf000064_0001
The one NT unit that developed a structure having greater than 16 cells was
plated down onto a fibroblast feeder layer. This structure was attached to the
feeder layer and started to propagate forming a colony with a ES cell-like mor¬
phology (See, e.g., Figure 2). Moreover, although the 4 to 16 cell stage structures
were not used to try and produce an ES cell colony, it has been previously shown
that this stage is capable of producing ES or ES-like cell lines (mouse, Eistetter et
al., Devel. Growth and Differ., 31:275-282 (1989); Bovine, Stice et al., 1996)).
Therefore, it is expected that 4 - 16 cell stage NT units should also give rise to
embryonic or stem-like cells and cell colonies.
Also, similar results were obtained upon fusion of an adult human
keratinocyte cell line with an enucleated bovine oocyte, which was cultured in
media comprising ACM, uridine, glucose, and 1000 IU of LIF. Out of 50
reconstructed embryos, 22 cleaved and one developed into a blastocyst at about day 12. This blastocyst was plated and the production of an ES cell line is
ongoing.
EXAMPLE 2
A. Mitochondria Isolation Protocol from a Cell
This Example relates to isolation of mitochondria and use thereof to
enhance the efficiency of cross-species nuclear transfer. The number of
mitochondria per cell varies from cell line to cell line. For example, mouse L cells
contain ~100 mitochondria per cell, whereas there are at least twice that number
in HeLa cells. The cells are swollen in a hypotonic buffer and ruptured with a few
strokes in a Dounce homogenizer using a tight-fitting pestle, and the mitochondria
are isolated by differential centrifugation.
The solutions, tubes, and homogenizer should be pre-chilled on ice. All
centrifugation steps are at 40°C. This protocol is based on starting with a washed
cell pellet of 1-2 ml. The cell pellet is resuspended in 11 ml of ice-cold RSB and
transferred to a 16 ml Dounce homogenizer.
RSB Buffer
RSB (A hypotonic buffer for swelling the tissue culture cells)
lO mM NaCl
1.5 mM MgCl2
Figure imgf000065_0001
MgCl2 The cells are allowed to swell for five to ten minutes. The progress of the
swelling is maintained using a phase contrast microscope. The swollen cells are
replaced, preferably by several strokes with a pestle. Immediately after, 8 ml of
2.5x MS buffer are added to give a final concentration of lx MS. The top of the
homogenizer is then covered with Parafilm and mixed by inverting a couple of
times.
2.5x MS Buffer
525 mM mannitol
175 Mm sucrose
Figure imgf000066_0001
215 mM EDTA pH 7.5
lx MS Buffer
210 mM mannitol
70 mM Sucrose
Figure imgf000066_0002
1 mM EDTA, pH 7.5
MS Buffer is an iso-osmotic buffer to maintain the tonicity of the
organelles and prevent agglutination.
Thereafter, the homogenate is transferred to a centrifuge tube for
differential centrifugation. The homogenizer is rinsed with a small amount of MS
buffer and added to the homogenate. The volume is brought to 30 ml with MS
buffer. The homogenate is then centrifuged at 1300 g for five minutes to remove nuclei, unbroken cells, and large membrane fragments. The supernatant is then
poured into a clean centrifuge tube. The nuclear spin-down is repeated twice. The
supernatant is then transferred to a clean centrifuge tube and a pellet containing the
mitochondria is centrifuged at 17,000 g for 15 minutes. The supernatant is
discarded and the inside of the tube wiped with a Kimwipe. The mitochondria is
washed by re-suspending the pellet in IX MS and repeating the 17,000 g
sedimentation. The supernatant is discarded and the pellet is resuspended in a
buffer. Mitochondria can be stored at -80°C for prolonged periods, e.g., up to a
year, but preferably will be used shortly thereafter for NT.
This basic protocol can be modified. In particular, it may be desirable to
further isolate mitochondrial DNA and us same for NT. In such case,
contamination with nuclei, not small organelles, potentially is a problem and the
following modifications may be made. For example, the cells may be harvested
in stationary growth phase when the fewest cells are actively dividing, and CaCl2
substituted for MgCl2 in the RSB to stabilize the nuclear membrane. The washing
of the mitochondrial pellet is omitted as is the density gradient purification.
Instead, the mitochondrial pellet is simply resuspended and lysed, and the
mitochondrial DNA purified from any remaining nuclear DNA. As noted, suitable
methods for purifying mitachondria and mitochondrial DNA are well known in the
art. Homogenization works best if the cells are resuspended in at least 5-1 OX
the volume of the cell pellet and if the cell suspension fills the homogenizer at
least half full. Press the homogenizer pestle straight down the tube, maintaining
a firm, steady pressure. The Dounce homogenizer disrupts swollen tissue culture
cells by pressure change. As the pestle is pressed down, pressure around the cell
increases; when the cell slips past the end of the pestle, the sudden decrease in
pressure causes the cell to rupture. If the pestle is very tight fitting, there may be
some mechanical breakage as well.
B. Isolation of Mitochondria From Tissue
A mitochondrial isolation protocol is selected based on the particular tissue.
For example, the homogenization buffer should be optimized for the tissue, and
the optimal way to homogenize the tissue utilized. Suitable methods are well
known in the art.
Rat liver is the most frequently used tissue for mitochondrial preparations
because it is readily available, is easy to homogenize, and the cells contain a large
number of mitochondria (1000-2000 per cell). For example, a motor-driven,
Teflon and glass Potter-Elvehjem homogenizer can be used homogenize rat liver.
Alternatively, if the tissue is soft enough, a Dounce homogenizer with a loose
pestle can be used. The yield and purity of the mitochondrial preparation is
influenced by the method of preparation, speed of preparation, and the age and physiological condition of the animal. As noted, methods of purifying
mitochondria are well known.
Preferably, the buffer, tubes, and homogenizer will be pre-chilled. Pre-
chilling a glass and Teflon type homogenizer creates the proper gap between the
tube and pestle. The centrifugation steps are preferably effected at 40°C.
Essentially, the process will comprise removal of the liver, taking care not
to rupture the gall bladder. This is placed in a beaker on ice and any connective
tissue is removed. The tissue is recognized and returned to the beaker, e.g., using
very sharp scissors, a scalpel, or razor blade, mince it into 1-2 slices. The pieces
are then rinsed, preferably twice, with homogenization buffer (IX MS) to remove
most of the blood, and the tissue transferred to the homogenizer tube. Enough
homogenization buffer if added to prepare a 1 : 10 (w/v) homogenate.
Use of Isolated Mitochondria or Mitochondrial DNA to Enhance NT
Efficacy
It is theorized by the inventors that the efficacy of cross-species nuclear
transfer may be enhanced by introduction of mitochondria or mitochondrial DNA
at the same species as donor cell or nucleus. Thereupon, the nucleus DNA of
resultant NT units will be species compatible.
Mitochondria isolated by the above or other known procedures are
incorporated, typically by injection, into any of the following (in the case of human
donor cell/bovine oocyte nuclear transfer): (i) non-activated, non-enucleated bovine oocytes;
(ii) non-activated, enucleated bovine oocytes;
(iii) activated, enucleated bovine oocytes;
(iv) non-activated, fused (with human donor cell or nucleus) bovine
oocytes;
(v) activated, fused and cleaved reconstructed (cow oocyte/human cell)
embryo; or
(vi) activated, fused one cell reconstructed (cow oocyte/human cell)
embryo.
The same procedures will enhance other cross-species NTs. Essentially,
mitochondria will again be introduced into any of (i)-(vi) of the same species as
the donor cell or nucleus, and the oocyte will be of a different species origin.
Generally about 1 to 200 picoliters of mitochondrial suspension are injected into
any of the above. The introduction of such mitochondria will result in NT units
wherein the mitochondrial and donor DNA are compatible.
EXAMPLE 3
Another method for improving the efficacy of the cross-species nuclear
transfer comprises the fusion of one or more enucleated somatic cells, typically
human (of the same species as donor cell or nucleus), with any of the following:
(i) non-activated, non-enucleated (e.g., bovine) oocyte;
(ii) non-activated, enucleated (e.g., bovine) oocyte; (iii) activated, enucleated (e.g., bovine) oocyte;
(iv) non-activated, fused (with human cell) oocyte (typically bovine);
(v) activated, fused and cleaved reconstructed (e.g., cow oocyte/human
cell) embryo;
(vi) activated, fused one cell reconstructed (cow oocyte/human cell)
embryo; or
(vii) non-activated, fused (e.g., with human cell) oocyte (typically bovine
oocyte).
Fusion is preferably effected by electrical pulse or by use of Sendai virus.
Methods for producing enucleated cells (e.g., human cells) are known in the art.
A preferred protocol is set forth below.
Enucleation Procedures:
Methods for the large-scale enucleation of cells with cytochalasin B are
well known in the art. Enucleation is preferably effected using the monolayer
technique. This method uses small numbers of cells attached to the growth surface
of a culture disc and is ideal if limited numbers of donor cells are available.
Another suitable procedure, the gradient technique, requires centrifugation of cells
through Ficoll gradients and is best suited for enucleation of large number (>107)
of cells.
Monolayer Technique. The monolayer technique is ideal for virtually any
cell which grows attached to the growth surface. Polycarbonate or polypropylene 250-ml wide-mouth centrifuge bottles with
screw-top caps are sterilized by autoclaving. The caps preferably are autoclaved
separately from the bottle to prevent damage to the centrifuge bottle. The bottle
are prepared for the enucleation procedure by the sterile addition of 30 ml DMEM,
2 ml bovine serum, and 0.32 ml cytochalasin B (1 mg/ml) to each. The caps are
placed on the bottles, and the bottles are maintained at 37° prior to use.
The cells to be enucleated (from a few hundred to ~105 cells) are seeded on
a culture dish (35 x 15 mm; Nunc Inc., Naperville, IL). Typically, the cells are
grown for at least twenty-four hours on the dishes to promote maximal attachment
to the growth surface. Preferably, the cells are prevented from becoming
confluent. The culture dish is prepared for centrifugation by wiping the outside
of the bottom half of the dish (containing the cells) with 70% (v/v) ethanol for the
purpose of sterilization. Alternatively, the dish can be kept sterile during cell
culturing by maintaining it within a larger, sterile culture dish. The medium is
removed from the dish and the dish (without top) is placed upside down within the
centrifuge bottle.
The rotor (GSA, DuPont, Wilmington, DE) and centrifuge are preferably
pre-warmed to 37° by centrifugation for 30-45 minutes at 8000 rpm. The HS-4
swinging-bucket rotor (DuPont) can alternatively be used. The optimal time and
speed of centrifugation varies for each cell type. For myoblasts and fibroblasts,
the centrifuge bottle with the culture dish is placed in the pre-warmed rotor and centrifuged for approximately 20 minutes (interval between the time when the
rotor reaches the desired speed and the time when the centrifuge is turned off).
Preferably, speeds of 6500 to 7200 rpm are used.
After centrifugation, the centrifuge bottle is removed from the rotor, and
the culture plate is removed from the bottles with forceps. A small amount of
medium is maintained in the plate to keep the cells moist in order to maintain cell
viability. The outside of the dish, including the top edge, is wiped with a sterile
wiper, then moistened with 95% (v/v) ethanol, to remove any medium and to dry
it. A sterile top is placed onto the dish. If the enucleated cells are not going to be
used immediately, complete culture medium (medium supplemented with the
appropriate concentration of serum) should be added to the dish, and the cells
placed in a C02 incubator. The resultant enucleated cells (karyoplast) are fused
with any of (i) - (viii) above.
While the present invention has been described and illustrated herein by
reference to various specific materials, procedures, and examples, it is understood
that the invention is not restricted to the particular material, combinations of
materials, and procedures selected for that purpose. Numerous variations of such
details can be implied and will be appreciated by those skilled in the art.

Claims

WHAT IS CLAIMED IS:
1. A method of producing embryonic or stem-like cells comprising the
following steps:
(i) inserting a desired differentiated human or mammalian cell
5 or cell nucleus into an enucleated animal oocyte, wherein such oocyte is derived
from a different animal species than the human or mammalian cell under condi¬
tions suitable for the formation of a nuclear transfer (NT) unit;
(ii) activating the resultant nuclear transfer unit;
(iii) culturing said activated nuclear transfer unit until greater than
0 the 2-cell developmental stage; and
(iv) culturing cells obtained from said cultured NT units to obtain
embryonic or stem-like cells.
2. The method of Claim 1 , wherein the cell inserted into the enucleated
5 animal oocyte is a human cell.
3. The method of Claim 2, wherein said human cell is an adult cell.
4. The method of Claim 2, wherein said human cell is an epithelial cell,
o keratinocyte, lymphocyte or fibroblast.
5. The method of Claim 2, wherein the oocytes are obtained from a
mammal.
6. The method of Claim 5, wherein the animal oocyte is obtained from
an ungulate.
7. The method of Claim 6, wherein said ungulate is selected from the
group consisting of bovine, ovine, porcine, equine, caprine, and buffalo.
8. The method of Claim 1 , wherein the enucleated oocyte is matured
prior to enucleation.
9. The method of Claim 1 , wherein the fused nuclear transfer units are
activated in vitro.
10. The method of Claim 1 , wherein the activated nuclear transfer units
are cultured on a feeder layer culture.
11. The method of Claim 10, wherein the feeder layer comprises fibro-
blasts.
12. The method of Claim 1, wherein in step (iv) cells from a NT unit
having 16 cells or more are cultured on a feeder cell layer.
13. The method of Claim 12, wherein said feeder cell layer comprises
fibroblasts.
14. The method of Claim 13, wherein said fibroblasts comprise mouse
embryonic fibroblasts.
15. The method of Claim 1, wherein the resultant embryonic or stem-
like cells are induced to differentiate.
16. The method of Claim 2, wherein the resultant embryonic or stem¬
like cells are induced to differentiate.
17. The method of Claim 1 , wherein fusion is effected by electrofusion.
18. Embryonic or stem-like cells obtained according to the method of
Claim 1.
19. Human embryonic or stem-like cells obtained according to the
method of Claim 2.
20. Human embryonic or stem-like cells obtained according to the
method of Claim 3.
21. Human embryonic or stem-like cells obtained according to the
method of Claim 4.
22. Human embryonic or stem-like cells obtained according to the
method of Claim 6.
23. Human embryonic or stem-like cells obtained according to the
method of Claim 7.
24. Differentiated human cells obtained by the method of Claim 16.
25. The differentiated human cells of Claim 24, which are selected from
the group consisting of neural cells, hematopoietic cells, pancreatic cells, muscle
cells, cartilage cells, urinary cells, liver cells, spleen cells, reproductive cells, skin
cells, intestinal cells, and stomach cells.
26. A method of therapy which comprises administering to a patient in
need of cell transplantation therapy isogenic differentiated human cells according
to Claim 24.
27. The method of Claim 26, wherein said cell transplantation therapy
is effected to treat a disease or condition selected from the group consisting of
Parkinson's disease, Huntington's disease, Alzheimer's disease, ALS, spinal cord
defects or injuries, multiple sclerosis, muscular dystrophy, cystic fibrosis, liver
disease, diabetes, heart disease, cartilage defects or injuries, burns, foot ulcers,
vascular disease, urinary tract disease, AIDS and cancer.
28. The method of Claim 26, wherein the differentiated human cells are
hematopoietic cells or neural cells.
29. The method of Claim 26, wherein the therapy is for treatment of
Parkinson's disease and the differentiated cells are neural cells.
30. The method of Claim 26, wherein the therapy is for the treatment of
cancer and the differentiated cells are hematopoietic cells.
31. The differentiated human cells of Claim 24, which contain and
express an inserted gene.
32. The method of Claim 1, wherein a desired gene is inserted, removed
5 or modified in said embryonic or stem-like cells.
33. The method of Claim 32, wherein the desired gene encodes a
therapeutic enzyme, a growth factor or a cytokine.
o
34. The method of Claim 32, wherein said embryonic or stem-like cells
are human embryonic or stem-like cells.
35. The method of Claim 32, wherein the desired gene is removed,
modified or deleted by homologous recombination.
5
36. The method of Claim 1, wherein the donor cell is genetically
modified to impair the development of at least one of endoderm, ectoderm and
mesoderm.
0 37. The method of Claim 1, wherein the donor cell is genetically
modified to increase differentiation efficiency.
38. The method of Claim 36, wherein the cultured nuclear transfer unit
is cultured in a media containing at least one capsase inhibitor.
39. The method of Claim 1, wherein the donor cell expresses a
detectable label that is indicative of the expression of a particular cyclin.
40. The method of Claim 36, wherein the donor cell has been modified
to alter the expression of a gene selected from the group consisting of SRF,
MESP-1, HNF-4, beta-1, integrin, MSD, GATA-6, GATA-4, RNA helicase A, and
H beta 58.
41. The method of Claim 37, wherein said donor cell has been
genetically modified to introduce a DNA that provides for expression of the Q7
and or Q9 genes.
42. The method of Claim 41, wherein said gene or genes are operably
linked to a regulatable promoter.
43. The method of Claim 1 , wherein the donor cell has been genetically
modified to inhibit apoptosis.
44. The method of Claim 43, wherein reduced apoptosis is provided by
altering expression of one or more genes selected from the group consisting of
Bad, Bok, BH3, Bik, Blk, Hrk, BNJP3, GimL, Bid, EGL-1, Bcl-XL, Bcl-w, Mcl-l,
Al, Nr-13, BHRF-1, LMW5-HL, ORF16, Ks-Bcl-2, E1B-19K, and CED-9.
45. The method of Claim 44, wherein at least one of said genes is
operably linked to an inducible promoter.
46. A mammalian somatic cell that expresses a DNA that encodes a
detectable marker, the expression of which is linked to a particular cyclin.
47. The cell of Claim 46, wherein the cyclin is selected from the group
consisting of cyclin Dl, D2, D3, BI, B2, E, A and H.
48. The cell of Claim 46, wherein the detectable marker is a fluorescent
polypeptide.
49. The cell of Claim 48, wherein said mammalian cell is selected from
the group consisting of human, primate, rodent, ungulate, canine, and feline cells.
50. The cell of Claim 48, wherein said cell is a human, bovine or
primate cell.
PCT/US2000/025090 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions WO2001019977A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
MXPA02002744A MXPA02002744A (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions.
AU77019/00A AU7701900A (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
JP2001523749A JP2003525031A (en) 1999-09-14 2000-09-14 Method for enhancing embryo development by genetically modifying embryonic cells or stem-like cell lines and donor cells produced by hybrid nuclear transfer or tissue culture conditions
CA002384413A CA2384413A1 (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
IL14854700A IL148547A0 (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
NZ517609A NZ517609A (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
BR0013999-8A BR0013999A (en) 1999-09-14 2000-09-14 Embryonic or stem cell-like cell lines produced by cross-species nuclear transplantation and methods to improve embryonic development by genetic alteration of donor cells or by tissue culture conditions
EP00966717A EP1214404A4 (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
IL148547A IL148547A (en) 1999-09-14 2002-03-06 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US39536899A 1999-09-14 1999-09-14
US09/395,368 1999-09-14

Publications (2)

Publication Number Publication Date
WO2001019977A1 WO2001019977A1 (en) 2001-03-22
WO2001019977A9 true WO2001019977A9 (en) 2002-09-26

Family

ID=23562752

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/025090 WO2001019977A1 (en) 1999-09-14 2000-09-14 Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions

Country Status (10)

Country Link
EP (1) EP1214404A4 (en)
JP (1) JP2003525031A (en)
CN (1) CN1379814A (en)
AU (1) AU7701900A (en)
BR (1) BR0013999A (en)
CA (1) CA2384413A1 (en)
IL (2) IL148547A0 (en)
MX (1) MXPA02002744A (en)
NZ (1) NZ517609A (en)
WO (1) WO2001019977A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7696404B2 (en) 1996-08-19 2010-04-13 Advanced Cell Technology, Inc. Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
WO2001018193A1 (en) * 1999-09-07 2001-03-15 Advanced Cell Technology, Inc. Method for generating immune-compatible cells and tissues using nuclear transfer techniques
CN1413250A (en) * 1999-10-15 2003-04-23 先进细胞技术公司 Method of producing differentiated progenitor cells and lineage-defective embryonic stem cells
CN1425064A (en) * 1999-12-20 2003-06-18 马萨诸塞大学 Embryonic or stem-like cells produced by cross species nuclear transplantation
US10865383B2 (en) 2011-07-12 2020-12-15 Lineage Cell Therapeutics, Inc. Methods and formulations for orthopedic cell therapy

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
EP0934403A4 (en) * 1996-08-19 2001-03-14 Univ Massachusetts Embryonic or stem-like cell lines produced by cross species nuclear transplantation
AU8587598A (en) * 1997-07-26 1999-02-16 Wisconsin Alumni Research Foundation Trans-species nuclear transfer
AU759322B2 (en) * 1998-03-02 2003-04-10 University Of Massachusetts Embryonic or stem-like cell lines produced by cross-species nuclear transplantation
CN1425064A (en) * 1999-12-20 2003-06-18 马萨诸塞大学 Embryonic or stem-like cells produced by cross species nuclear transplantation

Also Published As

Publication number Publication date
BR0013999A (en) 2002-05-21
EP1214404A4 (en) 2003-09-03
JP2003525031A (en) 2003-08-26
MXPA02002744A (en) 2003-07-21
CA2384413A1 (en) 2001-03-22
AU7701900A (en) 2001-04-17
WO2001019977A1 (en) 2001-03-22
EP1214404A1 (en) 2002-06-19
IL148547A (en) 2009-09-01
CN1379814A (en) 2002-11-13
IL148547A0 (en) 2002-09-12
NZ517609A (en) 2004-02-27

Similar Documents

Publication Publication Date Title
AU740709B2 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplanta tion
AU742363B2 (en) Nuclear transfer with differentiated fetal and adult donor cells
US20010012513A1 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation
EP1159404A2 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation
US7696404B2 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
NZ502129A (en) Cloning using donor nuclei from non-serum starved, differentiated cells
AU784731B2 (en) Embryonic or stem-like cells produced by cross species nuclear transplantation
AU759322B2 (en) Embryonic or stem-like cell lines produced by cross-species nuclear transplantation
AU2008229989A1 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
WO2001019977A9 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
US20050095704A1 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation
AU2005225103A1 (en) Embryonic or stem-like cell lines produced by cross species nuclear transplantation and methods for enhancing embryonic development by genetic alteration of donor cells or by tissue culture conditions
AU2006201208A1 (en) Embryonic or stem-like cells produced by cross species nuclear transpantation
MXPA00008602A (en) Embryonic or stem-like cell lines produced by cross-species nuclear transplantation

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 517609

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 148547

Country of ref document: IL

Ref document number: 77019/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2384413

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/002744

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2001 523749

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2000966717

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 008142785

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2000966717

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 10070719

Country of ref document: US

AK Designated states

Kind code of ref document: C2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 1-67, DESCRIPTION, REPLACED BY NEW PAGES 1-72; PAGES 68-75, CLAIMS, REPLACED BY NEW PAGES 73-81; PAGES 1/3-3/3, DRAWINGS, REPLACED BY NEW PAGES 1/3-3/3; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

WWP Wipo information: published in national office

Ref document number: 517609

Country of ref document: NZ

WWW Wipo information: withdrawn in national office

Ref document number: 2000966717

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 517609

Country of ref document: NZ