WO2002067669A2 - Production d une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale - Google Patents

Production d une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale Download PDF

Info

Publication number
WO2002067669A2
WO2002067669A2 PCT/US2002/000429 US0200429W WO02067669A2 WO 2002067669 A2 WO2002067669 A2 WO 2002067669A2 US 0200429 W US0200429 W US 0200429W WO 02067669 A2 WO02067669 A2 WO 02067669A2
Authority
WO
WIPO (PCT)
Prior art keywords
pgcs
gonadal
cell
cells
egg
Prior art date
Application number
PCT/US2002/000429
Other languages
English (en)
Other versions
WO2002067669A3 (fr
Inventor
Alexander Baguisi
Karl M. Ebert
Original Assignee
Tranxenogen, Inc.
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 Tranxenogen, Inc. filed Critical Tranxenogen, Inc.
Priority to JP2002567051A priority Critical patent/JP2005507234A/ja
Priority to CA002438612A priority patent/CA2438612A1/fr
Priority to EP02723043A priority patent/EP1361788A2/fr
Publication of WO2002067669A2 publication Critical patent/WO2002067669A2/fr
Publication of WO2002067669A3 publication Critical patent/WO2002067669A3/fr

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
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0608Germ cells
    • C12N5/0611Primordial germ cells, e.g. embryonic germ cells [EG]
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/30Bird
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • 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
    • C12N2510/00Genetically 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

Definitions

  • the invention relates to methods of genetically manipulating an avian genome. Extensive effort has been directed at developing better and more efficient methods for producing transgenic animals for commercial and research purposes. Recent developments in the field described the use of intracytoplasmic sperm injection (ICSI) for sperm-mediated transgenesis (Perry et al. 1999, Science 284: 1180-1183). Some researchers have reported the production of cloned animals by nuclear transfer in mammals, e.g., sheep (Wilmut etal.
  • ICSI intracytoplasmic sperm injection
  • the invention features an isolated avian gonadal cell, e.g., an ovarian cell or a testes cell, containing a heterologous nucleic acid.
  • the cell is preferably an embryonic cell.
  • avian refers to any avian species, including but not limited to, chicken, turkey, duck, goose, quail, and pheasant.
  • the invention also includes a method of introducing a nucleic acid molecule into the genome of an avian species, by contacting a population of isolated gonadal cells derived from a chick embryo with the nucleic acid molecule to yield transfected gonadal cells, and transferring the transfected gonadal cells to a fertilized avian egg.
  • the nucleic acid molecule is heterologous, i.e., it is derived from a breed or species which differs from the breed or species from which the gonadal cell is derived. Transfected gonadal cells are used then used to produce transgenic birds.
  • the population of isolated gonadal cells contains at least 0.5% primordial germ cells (PGCs), more preferably at least 1% primordial germ cells, and even more preferably at least 50% primordial germ cells.
  • PGCs primordial germ cells
  • Gonadal PGCs are isolated, i.e., separated, from other cells, e.g., stromal gonadal cells, with which they naturally-occur in a tissue.
  • the population is at least 80%, more preferably, 90%, more preferably 95%, and most preferably 99-100% gonadal PGCs.
  • the PGCs are transfected with heterologous DNA either before or after purification from other non-PGC cells.
  • Gonadal avian PGCs are cells that exist in an embryonic chick which can give rise to oocytes and sperm cells.
  • the chick embryo from which the gonadal PGCs are obtained is preferably at an embryonic stage of greater than 27 of the developing chick embryo (Hamburger &
  • the chick embryo is at an embryonic stage of 29-36 of development.
  • the chick embryo from which the gonadal cells are obtained has been incubated for at least 6.5 days, and more preferably 7.5 days (stage 29-36 of development) at the time of recovery.
  • Gonadal PGCs obtained from chick embryos at least 6.5 d but less than 15d of developmental age retain migratory capacity.
  • day 8, 9, 10, 11, 12, 13, or 14 gonadal PGCs are transferred to recipient eggs and retain migratory capability.
  • Transfected donor gonadal cells are derived from the same or different breed of bird compared to the breed from which the recipient egg is obtained.
  • the transfected donor gonadal PGCs are isolated from the same or different species of bird compared to the species from which the fertilized recipient avian egg is obtained.
  • the fertilized recipient avian egg is between stage 7-8 of development, i.e., the recipient egg has been incubated for at least 12 hours.
  • the fertilized avian egg is between stage 13-19 of development. Eggs which are unincubated include those which have just been laid or those which have been stored at a temperature less than 37°C.
  • unincubated eggs include those which have been stored at a storage temperature of approximately 60°F, at room temperature, or in the cold (e.g., at 4°C) from the time just after laying to the time of incubation.
  • Incubation refers to the time at which an egg is exposed to temperature conducive to development (e.g., 37-38°C or a temperature tailored to the requirements of the breed or species of bird).
  • the stage of development is determined chronologically or by visual examination of the state, e.g., size and morphology) of embryonic tissues.
  • Preferred promoters to regulate expression of heterologous sequences include tissue-specific promoters, e.g., those which direct expression of the transgene in oviductal cells of the chicken. Transgene expression in oviductal cells leads to accumulation of the trangene product in the albumen of the egg.
  • tissue-specific promoters e.g., those which direct expression of the transgene in oviductal cells of the chicken.
  • Transgene expression in oviductal cells leads to accumulation of the trangene product in the albumen of the egg.
  • the ovalbumin promoter known in the art is used to direct expression in oviductal cells.
  • a promoter directs expression of the transgene in liver tissue.
  • a vitellogenin promoter is used to direct expression of a heterologous polypeptide (encoded by the transgene) to the liver and secretion into the blood stream.
  • the transgene sequence operably linked to a vitellogenin promotoer encodes a light chain and heavy chain of an antibody, which confers disease resistance.
  • a vitellogenin promotoer encodes a light chain and heavy chain of an antibody, which confers disease resistance.
  • Introduction of such a construct is useful for breed improvement. Promoters which direct expression in blood, muscle, feathers, or other tissues are also used. Tissue- specific enhancer may also be used to augment expression in a preferred target tissue.
  • promoters which are not tissue-specific are used to direct expression systemically.
  • the transgene construct contains one or more promoters.
  • the construct contains two promoters, e.g., a first promoter operably linked to a sequence which encodes a transient marker gene (expression of which allows selection of transformed PGCs) and a second promoter which directs expression of the transgene in a tissue-specific manner.
  • transgenes include insulin and antibody molecules or fragments thereof.
  • the transgene encodes an intact heterodimeric monoclonal antibody, or an an immunologically-active antibody fragment, e. g. , a Fab or (Fab)2 fragment, an engineered single chain Fv molecule, or a chimeric molecule, e.g., an antibody which contains the binding specificity of one antibody, e.g., of murine origin, and the remaining portions of another antibody, e.g., of human origin.
  • Transgenes e.g., antibody encoding genes, the gene for human or porcine insulin (or insulin from another mammal) is operably linked to an ovalbumin promoter to allow accumulation of insulin in the egg white fraction of an egg.
  • a light chain encoding sequences are operably linked to an ovalbumin promoter, and heavy chain sequences are operably linked to an ovalbumin promoter for expression of both chains in oviductal cells.
  • the transgene product is then purified from the egg using methods known in the art.
  • Expression of antibody molecules is preferably targeted for accumulation in the egg white fraction of an egg (e.g., using an ovalbumin promotoer for expression in oviductal cells).
  • Donor PGCs are obtained from sex differentiated gonads and are therefore segregated by sex .
  • Donor PGCs stocks are sex-matched with the recipient egg. Such a strategy insures favorable fertility rates and high germline expression of the donor PGCs (tranfected or untransfected).
  • the sex of the recipient egg is hormonally controlled, e.g., by introducing testerone into the egg to generate a male chick or by introducing estrogen or follicle stimulating hormone into the egg to generate a female chick.
  • Untransfected PGCs of one species are used to generate birds of another species.
  • Transgenic birds in which a gene has been disrupted are also generated using the methods described above.
  • a knockout construct is made containing sequences complementary to sequences in the endogenous gene.
  • the sequences in the transgene construct undergo homologous recombination with the target sequences resulting in disruption of the gene. Disruption of the gene leads to production of a non-functional gene product or results in little or no production of the gene product. Genes which are involved in pathological conditions are disrupted in this manner.
  • the resulting knockout transgenic bird may be used as an animal model for the disease state.
  • Such knockout transgenic birds are produced using an isolated avian gonadal cell, which contains a genetic disruption of an endogenous gene, e.g., a disruption which inhibits production of a functional gene product.
  • the invention includes an avian egg containing a xenogeneic PGC, e.g., a gonadal
  • xenogeneic is meant that the PGC and the avian (recipient) egg are of different species.
  • a chicken egg contains a PGC from an emu or an avian species other than a chicken.
  • the invention also encompasses a chicken egg of one breed containing a gonadal PGC from another breed of chicken.
  • an isolated population of sex-determined PGCs e.g., a population of male gonadal PGCs or a population of female gonadal PGCs, is also within the invention.
  • the population of male gonadal PGCs contains less than 20%, preferably less than 10%, preferably less than 5%, and more preferably less than 1% female gonadal PGCs.
  • the population of female gonadal PGCs contains less than 20%, preferably less than 10%, preferably less than 5%, and more preferably less than 1% male gonadal PGCs.
  • Fig. 1 is a flow chart showing the procedure for isolating donor PGCs and preparing recipient eggs for transfer of donor PGCs.
  • Figs. 2 A and 2B are photographs of chicks generated from two pairs of breeding recipients that received donor PGCs.
  • Figs. 3 A and 3B are photographs of chicks generated from the transfer of both
  • Fig. 3A shows pure donor derived chicks generated from Rhode Island Red males bred to Barred White Rock females (commercially sold as Redi-Link Cross). The females are black and males are barred.
  • Fig. 3B shows pure donor derived chicks. Sex-linked chicks generated from the mating of Rhode Island Red males to Barred Rock females. The mating results in cockerel chicks with a white spot on the head and females do not (the popular commercial Red-Rock Cross). White chicks were derived from the endogenous germ cells of the White Leghorn recipients.
  • Fig. 3A shows pure donor derived chicks generated from Rhode Island Red males bred to Barred White Rock females (commercially sold as Redi-Link Cross). The females are black and males are barred.
  • Fig. 3B shows pure donor derived chicks. Sex-linked chicks generated from the mating of Rhode Island Red males to Barred Rock females. The mating results in cockerel
  • FIG. 4 is a photograph of a transgenic chick embryo showing expression of test transgene beta galactosidase.
  • PGCs were transfected with the transgene and transferred to a recipient egg. The recipient chick embryo was recovered at day 10 of incubation to test for transgene expression.
  • Beta galactosidase expression was detected in the mesonephros- gonadal region.
  • Donor PGCs were transfected in vitro using lipids and transferred to the germinal crescent region of a 48 h incubated egg that was partially sterilized with 75 micrograms of busulphan 24 h prior to PGC transfer. Beta galactosidase expression was under the control of the Cytomegalovirus promoter.
  • Avian gonadal cells are genetically engineered and used to introduce heterologous nucleic acids into the avian genome.
  • the methods are used to improve breed quality, produce avian models of non-avian diseases (e.g., human diseases), confer disease resistance to birds, and to produce recombinant proteins for pharmaceutical and other uses.
  • the use of gonadal cells to mediate DNA transfer allows the production of birds across the different breeds and between species of birds for conservation of endangered species using the chicken as the "universal recipient".
  • Isolated avian gonadal PGCs are used to produce transgenic animals in the avian species.
  • Heterologous nucleic acids are introduced into the PGCs to yield stable transformed germ cells.
  • the methods described herein are applicable to all birds, and not limited only to chickens, turkeys, pheasants, ducks and geese.
  • the overall efficiency of the procedure depends upon the timing of cell isolation, time and site of transfer, sex pre-selection of the donor PGCs, and the sterilization of the recipient embryo to maximize donor PGC population of the recipient gonads. Standard genetic engineering technologies are used to produce transgenic animals.
  • Donor cells are purified before or after nucleic acid transfer.
  • PGCs are transfected in vitro and then purified prior to transfer into a recipient egg.
  • the transfected (or transgenic gonadal PGCs are used to generate germline transgenic chickens for the production of pharmaceuticals in birds and eggs.
  • the sequence or structure of the transgene does not affect the success or efficiency of gene transfer.
  • the promoter which regulates expression of the transgene is benign or tissue specific.
  • Transgenic proteins or polypeptides are targeted for expression primarily in oviductal cells such that the gene product can be isolated from the egg after oviposition (laying of the egg).
  • tissue-specific promoters and/or enhancers allow expression in other tissues, including feathers, skin, and muscles.
  • the method can also be used for the generation of other avian species using the chicken /egg as the "universal recipient" for production of other bird species in chickens and conservation of endangered bird species.
  • the cells and methods described herein are useful to produce germline birds from donor PGCs.
  • the donor PGCs are transfected with heterologous nucleic acid and purified (or vice versa) so that an isolated population of transgenic PGCs are transferred to the recipient egg. Further enrichment of transfected PGCs is accomplished using a marker based cell sorter.
  • donor PGCs are transfected with a transgene that improves the quality of the bird or the meat and eggs the bird produces.
  • DNA encoding genes which confer disease resistance or a growth advantage are transferred to create animals with desirable market characteristics.
  • Therapeutic or pharmaceutical proteins or polypeptides are made by transfecting gonadal PGCs with DNA encoding the desired polypeptide, transferring the PGCs to a recipient egg, and incubating the egg under conditions suitable for the generation of germline chimera and subsequently transgenic birds for the production of the desired polypeptide.
  • Therapeutic proteins are targeted for expression in the yolk or albumen of the egg or in the meat, feathers, skin, blood and other parts of the bird.
  • the desired polypeptide is then isolated from the tissue or fluid (e.g., egg albumen) using methods known in the art.
  • the invention is also useful to produce a breed of bird which differs from the recipient breed (but is still within the same species, e.g., chicken).
  • the donor breed e.g., the same species, e.g., chicken.
  • PGCs are derived from a Rhode Island Red breed and transferred to an egg of another breed of chicken (e.g., a Madison Rock). Common American breeds of chickens include Madison Rock, Anthonys, Wyandottes, Rhode Island Reds, Rhode Island Whites, Buckeyes, Chanteclers, Jersey Giants, Lamonas, New Hampshires, and Delawares.
  • Use of isolated avian gonadal PGCs in non-trans genie applications Isolated but untransfected gonadal PGCs are used to produce cross-species birds. For example, a quail is produced from an egg generated by a chicken.
  • the methods of the invention provide for the use of a chicken or other domesticated bird as a "universal recipient" to produce cross-species birds for the conservation of endangered species.
  • the advantage of such a production approach is that since the chicken is a non-seasonal highly productive domesticated bird, endangered birds are reliably produced without the seasonal or geographic limitations associated with "endangered species.
  • the methods are used to produce rare or endangered bird such as the Houbarra Bustard, an endangered bird native to the Arabian peninsula.
  • Other endangered birds include pheasants, quails, parrots and macaws, all of which are threatened by habitat loss and exploitation by hunters and traders.
  • the methods are particularly useful to produce birds which are naturally seasonal breeders, e.g., pheasants or turkeys, or birds that do not breed in captivity.
  • Albatross and petrels which naturally breed on small oceanic islands, can also be reproduced by transfer of gonadal PGCs to a chicken recipient.
  • Many species of rails, cranes and kagus are also at high risk because they are very slow-breeding animals, making them extremely vulnerable to disturbance of nesting grounds and wintering areas.
  • Songbirds which account for almost 60 percent of all bird species, have a slightly below- average risk of extinction, but some species, including those in American grasslands, are in serious decline.
  • Example 1 Donor PGCs Freshly laid (or unincubated) fertilized eggs are incubated for up to 10 days in a humidified egg incubator at 37-38°C or at a temperature conducive to egg development. Egg incubation requirements for various species of birds is shown in Table 1.
  • Gonads were harvested from the eggs between days 6-10 (stage 29-36, Hamburger & Hamilton, 1951 J Morphol 88: 49-92). Alternatively, the gonads are harvested between days 7-8 (stage 31-34). The data indicate that from day 7 of incubation, morphological differences between the sexes were identifiable allowing sex selection (females have a bigger left gonad while males have similarly sized pair of gonads). The avian embryo has received neurotrophic signals for sex differentiation and exhibit differences between ovarian and testis development.
  • the PGCs retain migratory capacity and are remain responsive to chemoattractant factors which allow migration of the PGCs to the gonads of a developing embryo, e.g., in the recipient egg.
  • the gonads were grouped by sex and dispersed by standard trypsinization procedure and cultured in tissue culture plates until they attach. Sex selection is carried out at the time of collection of the gonadal PGCs prior to transfer of the cells into a recipient egg.
  • the PGCs can be transfected together with the other gonadal stromal cells or isolated prior to transfection using a ficoll density gradient (Yasuda etal, 1992, J. Reprod. Fert. 96: 521-528).
  • the gonadal stromal cells attach more rapidly to plastic, e.g., a tissue culture plate, than the PGCs, thus allowing purification of gonadal PGCs to approximately 90% or highter.
  • the gonadal PGCs are transfected using Lipids (1-2 ⁇ g/ml
  • the PGCs are transfected using calcium phosphate and other methods known to the art for introducing nucleic acid sequences.
  • the cells are transfected more than once to increase the transfection rate.
  • the PGCs are optionally cultured short term of up to 4 days in vitro without loss of migratory capability and without differentiation, allowing for several transfection repetitions and selection. Following short-term culture, the PGCs are separated from the stromal cells using standard methods. The transfected PGCs are further isolated by antibiotic selection, e.g., if the transgene construct carries a marker encoding antibiotic resistance.
  • FACS fluorescence activated cell sorter
  • magnetic cell sorting Other methods of cell purification include fluorescence activated cell sorter (FACS) isolation or magnetic cell sorting. These methods yield an isolated transgenic population of PGCs, which are then transferred to a recipient egg.
  • Donor gonadal PGCs are derived from the same or from a different breed of bird from the recipient egg. In the latter case, identification of donor PGC-produced germline chicks is facilitated by on a difference in distinguishing characteristics of the breed, e.g., color.
  • Example 2 Recipient Eggs Fertilized laid eggs are used as recipient eggs. Recipient eggs are incubated immediately or soon after laying or stored cold (unincubated) after laying but prior to transfer. Incubation begins upon exposure of the eggs to a warm temperature which is conducive to further development of the embryo (e.g., 37-38° C). A "0 hour” egg is one that is laid but has not yet been incubated. Similarly, a "12 hour” egg is one that has been incubated for 12 hours and so on. Fertilized eggs for recipients are used at 12, 24, 48, and up 96 hours from the start of incubation.
  • the developmental stage of the recipient egg is determined by chronologic age or hours post-incubation (see, e.g., Hamburger & Hamilton, 1951 J Morphol 88: 49-92) or by visual inspection of the stage of development of embyonic tissues.
  • PGCs are transferred to recipient eggs in a volume of 2-20 microliters. Approximately 50-1000 PGCs are transferred. For example, PGCs are transferred at a cell density of about 50-400 cells/microliter.
  • the site of transfer will depend on the developmental stage of the egg. At 0-12 hours of incubation, the site of transfer is into the blastodermal disc or into the blastocoel cavity. The volume in which donor PGCs are transferred is generally less than 5 microliters of medium. At 24 hours (stage 7-8, Hamburger & Hamilton, 1951 J Morphol
  • the site is off center (the margin between the area pellucida and the area opaca) so as not to physically affect the developing chick.
  • the site of transfer is at or near the germinal crescent region where PGCs normally congregate prior to entering the vasculature and migrating to the gonads.
  • the PGCs are injected straight into the vasculature at around 50-60 hours from the start of incubation or from stage 13-17 (Yasuda etal.,1992 J. Reprod and Fert 96:521-528, Naito, etal. 1998, J. Reprod and Fert ll3: 137-143).
  • PGCs are transferred to a recipient egg at stage 7-12 of development in which the blastocoelic cavity has been formed following short term incubation of the egg for 6-12 hours.
  • the PGCs are injected into the blastocoelic cavity.
  • the recipient egg is between stages 13-16 of development (egg incubation time of 24-28 hours, the PGCs are transferred in the region between the area pellucida and the area opaca.
  • the recipient egg is at a stage of development corresponding to 36-48 hours of incubation, the PGCs are injected directly into the germinal cresent region, where endogenous PGCs normally congregate prior to entering the chicken vasculature.
  • the recipient egg is at a stage of development corresponding to 50-72 hours (stage 16-19), the PGCs are transferred directly into the vasculature, e.g., into a blood vessel.
  • the egg is irradiated between 0-24 hours of incubation to eliminate the endogenous PGCs (Carsience etal 1993, Development 117: 669-675). Chemical sterilization using busulphan is also effective to eliminate endogenous PGCs (Aige-Gil & Simkiss 1991, Research in Veterinary Wscience 50: 139-144; Vick etal, 1993, J. Reprod and Fert 98: 637-641).
  • 50 ⁇ g of busulphan in 10-20 microlitters of media are injected in the germinal crescent region (where the PGCs accumulate prior to entering the vasculature).
  • the donor PGCs are transferred to the germinal crescent area at 48 hours of incubation or into the vasculature at a later time (between 55-72 hours).
  • Example 3 Improved Avian Transgenics Using Gonadal PGCs
  • Gonads were isolated as follows. Freshly laid fertilized chicken eggs were incubated for up to 8 days in a humidified egg incubator at 37-38 °C and between 85-88% relative humidity. Gonads were harvested from the developing chick embryo between days 4-8 (stage 29-36).
  • gonads are harvested between days 7-8 (stage 31-36) thereby maximizing the number of PGCs recovered with minimal loss of migratory capabilities when transferred back to recipient chick embryos.
  • the gonads were recovered by removing the mesonephros region from the abdominal cavity of the embryos and dissecting out the gonads from the mesonephros using fine tip forceps under low power magnification.
  • developmental differences between the differentiating female and the male gonads can be identified allowing for sex selection.
  • the developing pair of female gonads shows atrophy of the right gonad and an enlarging left gonad resulting from the differential colonization pattern of the germ cells.
  • the differentiating male gonads are similar in size and are distinctive from the female gonads. Although gonads can be isolated at later stages of development with more distinctive differences between the male and female gonads, the ability of isolated PGCs to migrate is reduced when transferred to recipient chick embryos.
  • Donor PGCs can be derived from the same breed or from a different breed of bird than the recipient egg. In the latter case, identification of donor PGC-produced germline chicks is facilitated by differences in distinguishing characteristics of the breed (e.g. feather color, size and skin pigmentation). It is preferable that the source of the PGCs pheno typically expresses a different feather color than the recipient chick embryo. This facilitates easier selection of chicks derived from the transferred PGCs. Isolation and culture of the germ cells The isolated gonads were grouped by sex and dispersed by standard trypsinization procedures. The PGCs were distinguished by their large size (12-20 microns) compared to the gonadal stromal cells.
  • the PGCs Under bright field microscopy, the PGCs contain numerous lipid droplets throughout the cytoplasm with the large nucleus occupying an eccentric location. Chicken PGCs have high glycogen content and thus are identifiable by periodic acid Schiff staining.
  • the PGCs can either be co-cultured with the gonadal stromal cells or separated prior to culture on plates at 37 °C in a humidified incubator with 5% CO 2 for a period of up to 4 days. In some cases, long term culture may result in spontaneous differentiation, loss of migratory capability and reduction of germ cell potential.
  • the PGCs were cultured in DMEM with high glucose content and supplemented with 10% FBS, 5% chicken serum and growth factors (basic Fibroblast Growth Factor, Insulin Growth Factor- 1 and Stem Cell Factor at 10 ⁇ g/ml and murine Leukemia Inhibitory Factor at 10 units/ml) to maintain their germ cell state.
  • the PGCs were separated from the gonadal stromal cells using a ficoll density gradient. For the gradient, a 1.5 ml centrifuge tube was sequentially layered with 0.5 ml each of 16% and 7% ficoll in PGC media and overlaid with a 0.2 ml gonadal cell suspension. The gradient was centrifuged at 800 x g for 30 min.
  • the PGC-rich fraction located between the 16% and 7 % gradient was aspirated, washed with PGC media and pelleted at 500 x g for 5 min. Further purification is accomplished by short-term culture (15-30 minutes) of the isolated cells allowing differential attachment of gonadal stromal cells to tissue culture plates while the PGCs remain in suspension. Transfection and selection of transgenic PGCs
  • the PGCs were transfected using lipids (i.e. Lipofectamine, according to vendors instructions) for 3-4 hours or by electroporation from 200-250 volts and between 750-950 microfarads with 20-50 ⁇ g/ml of DNA.
  • Two test transgenes were transfected. Using these methods, beta galactosidase under the control of the Cytomegalovirus promoter was used as a test transgene to track gonadal colonization patterns of the donor PGCs. Additionally, a human lactoferin promoter sequence was used. Recipient chick embryos were recovered at day 10 of incubation showing expression of the test transgene beta galactosidase in the mesonephros- gonadal region.
  • Donor PGCs were transfected in vivo using direct blastodermal disc injection prior to egg incubation, or in vitro using lipids. PGCs were transferred to the germinal crescent region of a 48 h incubated egg that was partially sterilized with 75 micrograms of busulphan 24 h prior to PGC transfer.
  • a recipient chick embryo was recovered at day 10 of incubation showing expression of the test transgene beta galactosidase in the mesonephros- gonadal region (Fig. 4).
  • Donor PGCs were transfected in vitro using lipids and transferred to the germinal crescent region of a 48 h incubated egg that was partially sterilized with 75 micrograms of busulphan 24 h prior to PGC transfer.
  • Beta galactosidase expression was under the control of the Cytomegalovirus promoter. A range 10 to 20% initial transfection rate was obtained using the above methods.
  • transfected PGCs were separated and purified by antibiotic selection if the transgene construct carries a marker encoding antibiotic resistance.
  • Other methods of cell purification can be used including fluorescence activated cell sorter (FACS) if a fluorescent marker is used for selection.
  • FACS fluorescence activated cell sorter
  • Fertilized eggs up to 7 days post oviposition were used as recipient eggs. Recipient eggs prior to, and up to 72 hours of incubation (up to stage 19), were utilized as PGC recipients. This time period includes the stages of chick development where the PGCs are actively migrating, and up to the time of localization in the primordial gonads.
  • PGCs at a concentration of 100-200 per microliter were injected using a glass micropipette (25-40 microns) attached to a Hamilton syringe under the control of a micrometer plunger. A volume of 5 microliters of PGCs suspended in culture medium was injected.
  • PGCs may be transferred to different targeted regions depending on the stage of development of the chick embryo: (a) into the subgerminal cavity of the blastodermal disc of fertile non-incubated eggs, (b) into the blastocoel cavity that separates the epiblast and the hypoblast of the developing embryo incubated for 6-12 hours (stage 3-4), (c) into the area pellucida adjacent to the developing embryo at 24-28 hours of incubation (stage 7-8), (d) into the germinal crescent region apical to the head process of the developing chick embryo incubated between 40-48 hours (stage 11-13), (e) 400-600 PGCs in 2-3 microliters of media are injected into the vasculature at 55-72 hours of incubation (stage 14-19) using a glass micropipette (40 microns outside diameter) attached to a micromanipulator.
  • the PGCs are injected into the dorsal aorta although larger marginal veins and arteries can be used as sites of injection. At this stage, PGCs circulate normally within the vascular system prior to migrating to the gonadal strom.
  • the circulating endogenous PGCs are physically removed from the bloodstream by aspirating the blood at the time when PGCs are in the vasculature, thus allowing partial sterilization or the egg irradiated between 0-24 hours of incubation to eliminate the endogenous PGCs.
  • Chemical sterilization using busulfan may also be used.
  • Busulfan is dissolved in Dimethylformamide and 75 ⁇ g in 50 microliters of sesame oil is injected into the yolk of recipient eggs 20- 24 hours after the start of incubation to reduce endogenous PGC involvement.
  • the donor PGCs were transferred to the germinal crescent region, 48 hours from the start of incubation or into the vasculature 55-72 hours from the start of incubation. Following transfer, the eggs were incubated at 37-38 °C in a humidified incubator until hatching.
  • the resulting germline chimeric bird is bred to the same breed as that from which the donor PGCs is derived.
  • This approach allows one to distinguish those birds derived from endogenous PGCs from those derived from donor PGCs.
  • the distinguishing phenotypic characteristics that can be attributed to a specific breed such as feather color allows easy identification of donor PGC derived chicks. For example, feather color identification of chicks indicated whether they were produced from donor derived PGCs (black feathers) or endogenous PGCs (white feathers).
  • the addition of a sex-linked trait further allows the selection of the males (e.g., white patch on the head) from the females.
  • FIG. 4 shows expression of a test transgene beta galactosidase.
  • Table 2 shows the results of PGC-mediated transfer of a second transgene, human lactoferrin.
  • Transfection of PGCs was carried out in vitro as described above or in vivo at day 0 of incubation (U.S.S.N. 09/587,128; hereby incorporated by reference). In vivo transfection was carried out by introducing DNA directly into the germinal disc of a first egg at day ) of incubation. The egg is then incubated for 7.5 days and the PGCs removed, cultured, and transferred to a second (recipient) egg. PGCs may be transfected again with transgene DNA in culture. Tranfected PGCs were then transferred to the germinal crest region (GCR) of a recipient egg. The recipient eggs were incubated, and day 14 chick embryonic tissues were tested for presence of the transgene by polymerase chain reaction (PCR).
  • PCR polymerase
  • An avian system allows expression of the gene product in the albumen or yolk fraction of the eggs or other tissues of the bird.
  • the avian species, specifically the chicken offers an inherent advantage over most if not all of the domestic livestock species currently available as target production systems. Its reproductive potential and short generation time is a potential advantage compared to the other species presently utilized for transgenic production.
  • the methods described herein offer an alternative assisted reproduction technology targeted towards the chicken where genetic modifications are directly targeted towards the germ cell population.
  • methods for foreign gene integration in the germ-line of birds is enhanced through technologies that improve the frequency of stable integration events. These include proper nucleic acid sequence construction with efficient vectors and promoters for targeted expression and improving gene delivery efficiency by multiple rounds of in vitro transfection of germ cells combined with methods to identify and isolate stable integrants prior to transfer to recipient chicken embryos.
  • the methods provide a system to produce and differentiate between chicks derived from donor PGCs or endogenous PGCs following partial sterilization.
  • the technology produced birds derived from donor PGCs with a high rate of germline transmission ranging from 25-78% where almost half of the chicks produced (49%) on average were donor derived.
  • Incorporating feather color as a visual marker additionally offers a mechanism to identify and differentiate donors from endogenous chicks, simplifying the production system.
  • the procedure eliminates the need to run DNA analysis on every chick produced following breeding. This saves time and resources thus reducing cost of producing germline transgenic chickens.
  • PGCs are extra-embryonic in origin and migration is a combination of active migration to the germinal crescent region, followed by a passive stage where they temporarily circulate in the vascular system prior to an active migration to the gonadal anlagen where PGC proliferation and gonadal sex differentiation occurs.
  • sex differentiation becomes morphologically evident between 7-9 d of embryonic development. The point of sex differentiation of other avian species is determined by visual inspection of the gonads.
  • This stage of gonadal development in chickens was targeted in order to: a) maximize the number of PGCs recovered, b) determine whether PGCs still retain migratory capacity at this stage where morphological differences in gonadal sex development is evident, c) determine the effects of breed dominance in germline transmission and d) study the effects of opposite sex transfer on the germline transmission of PGC donor-derived chicks.
  • a flow chart of the procedure is shown in Fig. 1. Gonads from 6.5-8.5 d embryos were isolated and morphologically separated by sex. PGCs were isolated by standard trypsinization procedures and mechanical disruption.
  • PGCs were co-cultured with their gonadal stromal cells at 37° C in 5 % CO 2 for 2 d in DMEM with high glucose in the presence of 10 % FBS, 5 % chicken serum supplemented with antibiotics and growth factors (bFGF, IGF-1, SCF at 10 ⁇ g/ml and mLIF at 10 U/ml).
  • the PGCs were isolated in a 1.5 ml centrifuge tube using a ficoll gradient sequentially layered with 0.5 ml each of 16 % and 7 % ficoll overlaid with 0.2 ml of cell suspension and spun for 30 min at 800 x g.
  • Germline transmission of donor-derived chicks ranged from 31-78 % between experimental groups with an average transmission rate of 49% (97/198) when parent stocks were bred together. Of this, 92% of the chicks were crosses and 8 % were pure donor derived chicks. When PGCs were derived from White Leghorn embryos and transferred to colored breeds, germline transmission rates were 47 % compared to 17 % when donor
  • Integrating the ability to introduce heterologous nucleic acid sequences into PGCs combined with efficient selection of stable integrants provide a platform technology for production of biopharmaceuticals. Furthermore, this reproductive technology is useful to improve production breeds, confer disease resistance, and increase production efficiency in avian agriculture. Additionally, PGC transfer is useful for avian conservation programs. For conservation purposes, the chicken, a non-seasonal highly productive bird, is used as a "universal recipient", providing alternative systems to enhance captive breeding programs for conservation year round.
  • avian PGCs are extra-embryonic in origin. At the time of oviposition in chickens, there are roughly about 50 PGCs interspersed within the 40-60 thousand cells comprising the blastodermal disc of a fertilized oviposited egg. Their migratory pattern follows a circuitous path involving a combination of active and passive migratory phases. During the first 24 hours of egg incubation, approximately 500 cells in the Area Pellucida of the blastodermal disc converges to establish the posterior end of the developing embryo. From the convergence, the primitive streak develops towards the anterior end in direct apposition to the area of convergence.
  • the PGCs actively migrate away from the convergence and towards the anterior end, independent of the developing embryo and congregating in the germinal crescent region above the head fold prior to entering the vasculature at about 2-2.5 days of incubation. Subsequently, the PGCs passively migrate within the circulatory system until they reach the genital ridge where they leave the vasculature and actively migrate and colonize the developing gonads.
  • Manipulations were targeted at a specific stage of embryonic development in chickens in which early morphological differences between the male and female gonads become evident.
  • the study was designed (a) to determine whether PGCs still retain migratory capacity at this stage of gonadal sex development; (b) to maximize the number of PGCs that can be recovered without loss of migratory capacity; (c) to determine whether different breeds exhibit breed dominance relative to germline transmission when donor PGCs are different from the breed of the recipient embryo; and (d) to determine whether transferring PGCs to recipients of similar or opposite sex have any influence in the rates of germline transmission.
  • Fertilized chicken eggs from Rhode Island Red, Barred Neighborhood Rock and White Leghorn were incubated up to 8 days in a humidified egg incubator at 37-38 °C and between 85-88% relative humidity. Gonads were isolated by harvesting the mesonephros from the genital ridge and dissecting out the gonads from the mesonephros using fine tip forceps under low power magnification. Between 7-7.5 days of incubation, the developing gonads in females exhibit a slightly larger left gonad compared to the right.
  • the differentiating male gonads on the other hand are similar in size providing the morphological distinction between the two sexes
  • the gonads were grouped based on breed (white or colored breeds) and by sex, and dispersed by standard trypsinization procedures.
  • the PGCs were co-cultured with the gonadal stromal cells in tissue culture plates at 37 °C in a humidified incubator with 5% CO 2 for up to 4 days.
  • the PGCs were cultured in DMEM as described in Example 3 above.
  • the PGCs which are larger (14-20 microns) compared to the other gonadal stromal cells, were separated using a ficoll density gradient as described in Example 3 above.
  • Gonadal cells were cultured in 4-well plates and fixed in 3.7% formaldehyde for 15 minutes and washed in PBS twice.
  • Chicken PGCs exhibit high levels of glycogen and are readily identified using Periodic Acid-Schiff staining.
  • the Periodic Acid-Schiff staining system supplied as a kit (Sigma Diagnostics) was used to differentiate PGCs from the stromal cells that comprise the rest of the gonads.
  • busulfan was optionally used to partially sterilize the recipients.
  • Busulfan was dissolved in Dimethylformamide and at 24 hours after the start of incubation, 75 ⁇ g of Busulfan in 50 ⁇ l of sesame oil was injected into the yolk of recipient eggs.
  • the donor PGCs were injected into the germinal crescent region apical to the head process of the developing recipient embryo.
  • a volume of 5-10 ⁇ l of PGCs (100-200 PGCs per ⁇ l) suspension was injected per egg using a 50-micron (OD) glass micropipette attached to a micrometer-controlled Hamilton syringe.
  • the eggs were sealed with parafilm and incubated at 37-38 °C in a humidified incubator for an additional 18 days and transferred to a hatching incubator to hatch.
  • a breeding pair randomly selected from each of the 6 experimental replicates was used as chimeric parent stocks.
  • White leghorn recipients with Rhode Island Red and Barred Rock donor PGCs were bred with each other, and Rhode Island Red recipients with White Leghorn Donor PGCs were bred together.
  • the birds were pen mated and eggs derived from each breeding replicate were hatched on a weekly basis.
  • First generation chicks were identified as donor-derived or endogenous PGC-derived based on the characteristic feather coloration attributed to the breed of the birds or the crosses derived from them.
  • Chi-Square method was used to analyzed the data at a significance level of ⁇ 0.05.
  • the average hatching rates of recipient eggs was 37% from a range of 9-67%.
  • First generation chicks identified by feather markings as being derived from donor PGCs ranged from 17-71% among the breeding groups with an average transmission rate of 49% (97/198. Data from additional hatchings are shown in Table 3.
  • the mating system using both male and female recipients as breeding pair increased the rates of transmission and also resulted in pure donor PGC-derived chicks.
  • the chicks identified as being donor-derived 85% were from crosses between donors and endogenous germ cells and 15 % were pure donor-derived chicks (Figs. 2A, 2B).
  • the breeding scheme using White leghorn recipient breeding pairs receiving Rhode Island Red and Barred Rock PGCs produced 3 crossbreds and 3 purebred strains. Furthermore, a sex-linked feather trait was observed when pure donor-derived chicks were obtained from a breeding combination of PGCs derived from Rhode Island Red and Barred Rock (Figs. 3 A, 3B).
  • Transgenic animals are produced by introducing heterologous nucleic acid sequences under the control of appropriate regulatory promoters directly into the germ cell population and selecting stable integrants prior to transferring transgenic PGCs into recipient eggs.
  • a significant advantage of this system is that it can be used to enhance flock management and reduce overall production cost by incorporating feather color to identify chicks derived from manipulated germ cells, and select them at day old using sex-linked feather color trait.
  • the PGC method also provides an alternative assisted reproduction technology to enhance avian conservation programs, because the chicken, a non-seasonal highly productive bird is used as a "universal recipient" providing alternative systems to enhance captive breeding programs for avian conservation year round.
  • Example 5 Avian PGCs as stem cells for somatic tissue generation Stem cells are the primordial units of embryonic generation and adult regeneration.
  • primordial germ cells In mammals, primordial germ cells (PGCs) have the ability to remain undifferentiated and continuously proliferate in culture as well as exhibit multipotent capacity to participate in the formation of the three embryonic germ layers.
  • PPCs primordial germ cells
  • PGCs and shows that chicken PGCs in the early stages of gonadal sex differentiation still retain stem cell potential and can differentiate spontaneously in vitro and contribute to various tissues of the developing embryos in vivo after embryonic lineage differentiation has been established. PGCs exhibited a tendency to differentiate towards the neural and mesenchymal lineages. Using a fluorescent transgene marker (DsRed) to provide an unambiguous tag, observable localization patterns and extent of multipotency and plasticity in vivo were established in the developing fetus. The data indicate that isolated PGCs from sexually differentiating 6.5-8.5 d chicken gonads had not committed to an irreversible somatic lineage and were still capable of contributing to all germ layers in vivo.
  • DsRed fluorescent transgene marker
  • vasculature in chickens is not limiting, but facilitates the wide spread localization of PGCs and their subsequent multilineage involvement in various tissues of the developing fetus.
  • the gonadal PGCs described herein retained the intrinsic capacity to respond to extrinsic signal regulators for multilineage differentiation.
  • Freshly laid fertilized Barred Rock eggs were incubated up to 8.5 days in a humidified egg incubator at 37-38 °C and between 85-88% relative humidity.
  • Gonads were harvested from the developing chick embryo between 7-8 days of incubation (stage 31-34).
  • the gonads were recovered by removing the mesonephros from the genital ridge and dissecting out the gonads from the mesonephros using fine tip forceps under low power magnification.
  • the gonads were grouped according to sex. Females have a larger left gonad compared to the right, while male gonads exhibit a similar size.
  • the PGCs were isolated following standard trypsinization procedures and cultured with or without the gonadal stromal cells in tissue culture plates at 37 °C in a humidified incubator with 5% CO 2 .
  • the PGCs were cultured and isolated using a ficoll density gradient as described above. Periodic Acid-Schiff staining was carried out as described above.
  • the PGCs were transfected with Ds Red (Clontech Laboratories) to express cytoplasmic red fluorescence.
  • Ds Red Clontech Laboratories
  • Lipofectamine (GIBCO-BRL) at 15 ⁇ l was diluted to a final volume of 10 ⁇ l in OPTI-MEM 1 (GIBCO-BRL) and 1 ⁇ g of linearized DNA (Ds Red) was also diluted to the same final volume in OPTI-MEM 1 (GIBCO-BRL).
  • the preparations were combined and incubated for 30 minutes at room temperature and then added drop- wise into the wells. Culture media was added after 4 hours to inhibit toxicity. Consecutive rounds of transfections were used to improve the rate of transfection.
  • the PGCs were removed from gonadal- stromal co-culture and induced to spontaneously differentiate in the absence of growth factor supplements.
  • Colonies proliferated over 3 months in repeated subcultures in the presence of mitotically active gonadal stromal cells. Multipotent germ cells were uniformly round and did not attach tightly to the stromal layers or to other PGCs. Colonies were multi-layered and well delineated from the stromal layers. Loss of monolayer support led to some level of spontaneous differentiation even in the presence of both inhibitory and growth factor supplements. Long-term culture also showed a decrease in Periodic Acid-Schiff staining intensity.
  • PGCs differentiated spontaneously into several cell lineages.
  • the majority of the PGC cultures differentiated into fibroblast-like and mesenchymal type cells. Extended cultures of these cells subsequently led to a variety of connective-tissue cell types morphologically similar to bone, cartilage, muscle and immature fat cells.
  • PGCs proliferated into small compacted clusters of cells with minimal or loose monolayer attachment they often differentiated into neural cells forming extensive network between them.
  • PGCs also differentiated occasionally into an epithelial monolayer at confluence.
  • Multipotency and plasticity in vivo Using the vascular system as the route of transfer facilitated extensive localization and lineage involvement of PGCs in the developing chicken fetus. They were consistently observed in the heart, lungs and blood; and were estimated based on Ds Red expression to have contributed up to 20, 5 and 40% of the heart, lungs, and blood respectively. They were also observed to localize in the midbrain, eyes, bone marrow, muscle, gonads, mesonephros and skin. Lineage involvement was evident by fluorescence in the muscle, skin, bone marrow, gonads, mesonephros and the midbrain at different levels. Feather color patterns of hatched chicks also showed some contribution from the germ cells.
  • Sub-cultured germ cells maintained in the absence of gonadal-stromal monolayer support showed a reduction in multilineage involvement in vivo, specifically loosing their capacity to incorporate into the gonadal involvement.
  • the data indicate that isolated PGCs from sexually differentiating 7-8.5 day chicken gonads have not committed to an irreversible somatic lineage towards spermatogenesis and oogenesis. These cells still retain the multipotent capacity to differentiate in vitro and exhibit plasticity to contribute into all germ layers in vivo.
  • the vascular system in chickens is not limiting, but facilitates the wide spread localization of PGCs, allowing them to contribute to tissue generation and organogenesis in the developing fetus. These cells can be harnessed and reprogrammed by extrinsic regulators controlling the environmental milieu that determines their lineage differentiation. Although humans do not share a similar basic body plan to avian or to other animal models used for stem cell research, homologous parts that likely arise from the same biochemical mechanisms are shared. Thus, the chicken PGC model is a useful alternative to other animal models for the development of applications of stem cells in tissue engineering, embryonic generation, adult regeneration and gene therapy.

Abstract

La présente invention se rapporte à une cellule gonadique aviaire isolée, par ex. une cellule ovarienne ou une cellule testiculaire, contenant un acide nucléique hétérologue, ainsi qu'à une méthode destinée à introduire une molécule d'acide nucléique dans le génome d'une espèce aviaire et qui consiste à mettre une population de cellules gonadiques isolées dérivées d'un embryon de poulet en contact avec la molécule d'acide nucléique afin de produire des cellules gonadiques transfectées, et à transférer les cellules gonadiques transfectées dans un ovocyte fécondé.
PCT/US2002/000429 2001-02-16 2002-01-11 Production d une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale WO2002067669A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2002567051A JP2005507234A (ja) 2001-02-16 2002-01-11 始原生殖細胞ベースの、鳥の生殖細胞系の作製
CA002438612A CA2438612A1 (fr) 2001-02-16 2002-01-11 Production d une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale
EP02723043A EP1361788A2 (fr) 2001-02-16 2002-01-11 Production d'une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US69442701A 2001-02-16 2001-02-16
US26944201P 2001-02-16 2001-02-16
US60/269,442 2001-02-16

Publications (2)

Publication Number Publication Date
WO2002067669A2 true WO2002067669A2 (fr) 2002-09-06
WO2002067669A3 WO2002067669A3 (fr) 2003-02-27

Family

ID=29250345

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/000429 WO2002067669A2 (fr) 2001-02-16 2002-01-11 Production d une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale

Country Status (4)

Country Link
EP (1) EP1361788A2 (fr)
JP (1) JP2005507234A (fr)
CA (1) CA2438612A1 (fr)
WO (1) WO2002067669A2 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007501625A (ja) * 2003-08-08 2007-02-01 アビコアー バイオテクノロジー インスティチュート インク 鳥類精原幹細胞の培養方法及びこれにより収得した鳥類精原幹細胞
EP1850659A2 (fr) * 2005-02-01 2007-11-07 Origen Therapeutics, Inc. Poulets transgeniques
CN1761756B (zh) * 2003-01-16 2013-03-13 北卡罗来纳州大学 禽类物种中内源性原生殖细胞的耗减
AU2012201824B2 (en) * 2005-02-01 2014-08-28 Synageva Biopharma Corp. Transgenic chickens
EP3090629A1 (fr) * 2005-02-01 2016-11-09 Synageva BioPharma Corp. Méthode pour produire des poulets transgéniques utilisant une culture à long terme de cellules germinales primordiales de poulet
CN111965094A (zh) * 2020-08-24 2020-11-20 扬州大学 一种比较体外诱导的PGC-like细胞迁移效率的方法

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106070010B (zh) * 2016-06-15 2018-11-16 上海市农业科学院 一种黑羽蛋鸡配套系父本的育种方法及其配套系的制种模式

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990011355A1 (fr) * 1989-03-17 1990-10-04 National Research Development Corporation Introduction d'un gene exogene dans des oiseaux

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990011355A1 (fr) * 1989-03-17 1990-10-04 National Research Development Corporation Introduction d'un gene exogene dans des oiseaux

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BAGUISI A ET AL: "Avian primordial germ cells: Potential for transgenesis and conservation." THERIOGENOLOGY, vol. 57, no. 1, 2 January 2002 (2002-01-02), page 775 XP002216115 Proceedings of the Annual Conference of the International Embryo Transfer Society;Foz do Iguassu, Parana, Brazil; January 12-15, 2001 ISSN: 0093-691X *
BRESLER M ET AL: "MANIPULATIONS OF GERM-CELL POPULATIONS IN THE GONAD OF THE FOWL" BRITISH POULTRY SCIENCE, LONGMAN GROUP, GB, vol. 35, May 1994 (1994-05), pages 241-247, XP000994671 ISSN: 0007-1668 *
EBERT K M ET AL: "Avian primordial germ cells: Potential for somatic lineage contribution." THERIOGENOLOGY, vol. 57, no. 1, 2 January 2002 (2002-01-02), page 564 XP002216114 Proceedings of the Annual Conference of the International Embryo Transfer Society;Foz do Iguassu, Parana, Brazil; January 12-15, 2001 ISSN: 0093-691X *
ETCHES R J ET AL: "STRATEGIES FOR THE PRODUCTION OF TRANSGENIC CHICKENS" METHODS IN MOLECULAR BIOLOGY, HUMANA PRESS INC., CLIFTON, NJ, US, vol. 62, 1997, pages 433-450, XP000974906 *
FURUTA H ET AL: "DEVELOPMENT OF THE GONADS DERIVED FROM HETERO-SEXUALLY TRANSFERRED PRIMORDIAL GERM CELLS (PGCS) BETWEEN EMBRYOS IN THE CHICKEN" ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES, SUWEON, KR, vol. 12, December 1999 (1999-12), pages 1188-1191, XP000994668 ISSN: 1011-2367 *
NAITO M ET AL: "Expression of exogenous DNA in the gonads of chimaeric chicken embryos produced by transfer of primordial germ cells transfected in vitro and subsequent fate of the introduced DNA." JOURNAL OF REPRODUCTION AND FERTILITY, vol. 113, no. 1, May 1998 (1998-05), pages 137-143, XP002216113 ISSN: 0022-4251 cited in the application *
ONO T ET AL: "SETTLEMENT OF QUAIL PRIMORDIAL GERM CELLS IN CHICKEN GONADS" ANIMAL FEED SCIENCE AND TECHNOLOGY, AMSTERDAM, NL, vol. 69, no. 6, June 1998 (1998-06), pages 546-555, XP000994649 *
PERRY M M ET AL: "TRANSGENESIS IN CHICKENS" TRANSGENIC RESEARCH, LONDON, GB, vol. 2, no. 3, May 1993 (1993-05), pages 125-133, XP000998723 ISSN: 0962-8819 *
VICK L ET AL: "TRANSGENIC BIRDS FROM TRANSFORMED PRIMORDIAL GERM CELLS" PROCEEDINGS OF THE ROYAL SOCIETY OF EDINBURGH. SECTION B, BIOLOGICAL SCIENCES, ROYAL SOCIETY OF EDINBURGH, EDINBURGH, GB, vol. 251, no. 1332, 22 March 1993 (1993-03-22), pages 179-182, XP000997980 ISSN: 0269-7270 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1761756B (zh) * 2003-01-16 2013-03-13 北卡罗来纳州大学 禽类物种中内源性原生殖细胞的耗减
JP2007501625A (ja) * 2003-08-08 2007-02-01 アビコアー バイオテクノロジー インスティチュート インク 鳥類精原幹細胞の培養方法及びこれにより収得した鳥類精原幹細胞
EP1850659A2 (fr) * 2005-02-01 2007-11-07 Origen Therapeutics, Inc. Poulets transgeniques
EP1850659A4 (fr) * 2005-02-01 2010-12-08 Origen Therapeutics Inc Poulets transgeniques
AU2012201824B2 (en) * 2005-02-01 2014-08-28 Synageva Biopharma Corp. Transgenic chickens
EP3090629A1 (fr) * 2005-02-01 2016-11-09 Synageva BioPharma Corp. Méthode pour produire des poulets transgéniques utilisant une culture à long terme de cellules germinales primordiales de poulet
CN111965094A (zh) * 2020-08-24 2020-11-20 扬州大学 一种比较体外诱导的PGC-like细胞迁移效率的方法
CN111965094B (zh) * 2020-08-24 2023-10-27 扬州大学 一种比较体外诱导的PGC-like细胞迁移效率的方法

Also Published As

Publication number Publication date
EP1361788A2 (fr) 2003-11-19
CA2438612A1 (fr) 2002-09-06
JP2005507234A (ja) 2005-03-17
WO2002067669A3 (fr) 2003-02-27

Similar Documents

Publication Publication Date Title
Tajima et al. Production of germ line chimera by transfer of primordial germ cells in the domestic chicken (Gallusdomesticus)
CHANG et al. Production of germline chimeric chickens by transfer of cultured primordial germ cells
US7145057B2 (en) Chimeric bird from embryonic stem cells
US6156569A (en) Prolonged culturing of avian primordial germ cells (PGCs) using specific growth factors, use thereof to produce chimeric avians
DE69832716T2 (de) Herstellung embryonischer vogel keimzellinien durch verlängerte kultivierung von pgcs, verwendung desselben zur klonierung und chimerisierung
Petitte et al. Accessing the genome of the chicken using germline chimeras
Naito et al. Production of quail‐chick chimaeras by blastoderm cell transfer
AU736087B2 (en) Avian primordial germ cell (PGC) cell line and a method for long term culturing thereof
Bednarczyk et al. Improvement of hatchability of chicken eggs injected by blastoderm cells
US20020162134A1 (en) Primordial germ cell-based germ line production of birds
EP1361788A2 (fr) Production d'une lignee germinale d'oiseaux fondee sur une cellule germinale primordiale
JP4376901B2 (ja) 精原細胞を利用した鳥類キメラの生産方法及び鳥類キメラ
AU2002253850A1 (en) Primordial germ cell-based germ line production of birds
KR100267633B1 (ko) 배양된 원시생식세포로 전이된 생식선 키메라 조류의 생산방법
WO2000008132A1 (fr) Culture prolongee de cellules germinales primordiales d'aviens au moyen de facteurs de croissance specifiques, et leur emploi
Mahdi et al. An overview of the basics of generate transgenic hen „bioreactors”
한재용 et al. Expression of Lac Z gene in young chick gonad by the transfected primordial germ cell injection
Mahdi et al. Overview on the efforts to generate transgenic chicken
Naito et al. Fate of the Donor Blastodermal Cells Derived from the Central Disc, Marginal Zone and Area Opaca Transferred into the Recipient Embryos
MXPA00001301A (en) Avian primordial germ cell (pgc) cell line and a method for long term culturing thereof
AU2003214983A1 (en) Chimeric bird from embryonic stem cells
MXPA00001300A (en) Production of avian embryonic germ (eg) cell lines by prolonged culturing of pgcs, use thereof for cloning and chimerization

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC 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 PH 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: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM 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 TR BF BJ CF CG CI CM GA GN GQ 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)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC 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 PH 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: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM 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 TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2002253850

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2002567051

Country of ref document: JP

Ref document number: 2438612

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002723043

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002723043

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2002723043

Country of ref document: EP