Evidence of in vivo transfection of circulating primordial germ cells in chickens

Similar documents
, Takahiro Tagami, Hiroshi Kagami and Tamao Ono

Avian Germplasm Preservation: Embryonic Stem Cells or Primordial Germ Cells? 1

Research Fellow of the Japan Society for the Promotion of Science, Japan

DIFFERENTIATION OF FEMALE PRIMORDIAL GERM CELLS IN THE MALE TESTES OF CHICKEN

Germline Replacement by Transfer of Primordial Germ Cells into Partially Sterilized Embryos in the Chicken 1

Germ Cell Transplantation in Fish

Effects of 17beta-estradiol on distribution of primordial germ cell migration in male chicks

c Tuj1(-) apoptotic live 1 DIV 2 DIV 1 DIV 2 DIV Tuj1(+) Tuj1/GFP/DAPI Tuj1 DAPI GFP

Supplementary Document

University of Science and Technology in Bydgoszcz. Faculty of Animal Breeding and Biology. Animal Biochemistry and Biotechnology Department

Production of Offspring from Cryopreserved Chicken Testicular Tissue 1

Supplementary Table 3. 3 UTR primer sequences. Primer sequences used to amplify and clone the 3 UTR of each indicated gene are listed.

Testicular germ cells can colonize sexually undifferentiated embryonic gonad and produce functional eggs in fish

Supplemental Data. Shin et al. Plant Cell. (2012) /tpc YFP N

Supplementary Appendix

a) Primary cultures derived from the pancreas of an 11-week-old Pdx1-Cre; K-MADM-p53

Plasmids Western blot analysis and immunostaining Flow Cytometry Cell surface biotinylation RNA isolation and cdna synthesis

Yusaku Kohara, Yukio Kanai and Atsushi Tajima

A Method for Cryopreserving Semen from Yakido Roosters using. N-Methylacetamide as a Cryoprotective Agent

Supplementary Figure 1. ROS induces rapid Sod1 nuclear localization in a dosagedependent manner. WT yeast cells (SZy1051) were treated with 4NQO at

Toluidin-Staining of mast cells Ear tissue was fixed with Carnoy (60% ethanol, 30% chloroform, 10% acetic acid) overnight at 4 C, afterwards

REPRODUCTIVE BIOTECHNOLOGY IN SWINE

*To whom correspondence should be addressed. This PDF file includes:

Figure S1. Analysis of genomic and cdna sequences of the targeted regions in WT-KI and

SUPPLEMENTARY DATA. Supplementary Table 1. Primer sequences for qrt-pcr

Isolation and Characterization of Gonadal Primordial Germ Cells (gpgcs) of Turkey (Meleagris gallopavo) from Days Old Embryos

Phylogenetic analysis of human and chicken importins. Only five of six importins were studied because

A smart acid nanosystem for ultrasensitive. live cell mrna imaging by the target-triggered intracellular self-assembly

SUPPLEMENTARY INFORMATION

CD31 5'-AGA GAC GGT CTT GTC GCA GT-3' 5 ' -TAC TGG GCT TCG AGA GCA GT-3'

Supplementary Figure 1

Supplementary Materials and Methods

Supplementary Figure 1 a

Nature Structural & Molecular Biology: doi: /nsmb Supplementary Figure 1

Recommended laboratory tests to identify influenza A/H5 virus in specimens from patients with an influenza-like illness

Abbreviations: P- paraffin-embedded section; C, cryosection; Bio-SA, biotin-streptavidin-conjugated fluorescein amplification.

II. The effect of strain and IGF-1 gene transfer using SMGT techniques on some semen characteristics:-

Supplementary Materials

Bi-potent Gonads. Sex Determination

Expression of Selected Inflammatory Cytokine Genes in Bladder Biopsies

Journal of Cell Science Supplementary information. Arl8b +/- Arl8b -/- Inset B. electron density. genotype

BIOLOGY 621 Identification of the Snorks

Strategic delivery: Setting standards Increasing and. Details: Output: Demonstrating efficiency. informing choice.

Preimplantation genetic diagnosis: polar body and embryo biopsy

SUMMARY. Keywords: quail, Coturnix japonica, morphology, ovary, oviduct, neurotrophins, immunohistochemistry

ice-cold 70% ethanol with gentle vortexing, incubated at -20 C for 4 hours, and washed with PBS.

Table S1. Oligonucleotides used for the in-house RT-PCR assays targeting the M, H7 or N9. Assay (s) Target Name Sequence (5 3 ) Comments

The development of ovary in quail s embryo

Supplementary Figure 1

Supplementary Figure 1 MicroRNA expression in human synovial fibroblasts from different locations. MicroRNA, which were identified by RNAseq as most

Supplementary Information

Supplementary Figures

Supplementary Table 2. Conserved regulatory elements in the promoters of CD36.

Oogenesis. Key Concepts. Female Reproductive Tract

BIOL2005 WORKSHEET 2008

Chapter 4 The Chromosome Theory of Inheritance

Characterizing intra-host influenza virus populations to predict emergence

MEASURING FERTILITY & EARLY DEADS LEVELS PART 1

Pakistan Veterinary Journal

Citation for published version (APA): Oosterveer, M. H. (2009). Control of metabolic flux by nutrient sensors Groningen: s.n.

Sex is determined by genes on sex chromosomes

Frequency of mosaicism points towards mutation prone early cleavage cell divisions.

Changes in Quail Blastodermal Cell Status as a Result of Selection*

Triploidy and other chromosomal

Formylpeptide receptor2 contributes to colon epithelial homeostasis, inflammation, and tumorigenesis

Hepatitis B Virus Genemer

Fresh and Frozen Ovary Tissue Transplants: Mechanism of Adult Primordial Follicle Recruitment And Fetal Oocyte Arrest

(FITC) or rhodamine blue isothiocyanate (RBITC) for use in mixed egg-transfer experiments. Both FITC and RBITC bind to the zona pellucida

Biology 4361 Developmental Biology. October 11, Multiple choice (one point each)

Temperature during the initial phases of incubation

Comparison between Low/Programmable Freezing and Fast Freezing Protocols of Hungarian Guinea Fowl Semen

Nature Immunology: doi: /ni.3836

Construction of a hepatocellular carcinoma cell line that stably expresses stathmin with a Ser25 phosphorylation site mutation

Beta Thalassemia Case Study Introduction to Bioinformatics

SUPPLEMENTARY INFORMATION

Spermatogonial Transplantation in Fish: Production of Trout Offspring from Salmon Parents

Monitoring of the effects of various spiking techniques in broiler breeder flocks by counting of IPVL holes of the eggs

Supplemental Information. Cancer-Associated Fibroblasts Neutralize. the Anti-tumor Effect of CSF1 Receptor Blockade

SUPPORTING ONLINE MATERIAL

Biology 4361 Developmental Biology Exam 1 ID#: October 11, 2005

EFFECTS OF SOME PESTICIDES ON THE EMBRYONIC STAGES OF CHICK

DOES in ovo ADMINISTRATION OF ZINC OR IODINE MODULATE DIFFERENTIAL EXPRESSION OF GROWTH AND IMMUNE RELATED GENES IN BROILER CHICKENS

Supplementary Figure 1

Supplemental Information. Th17 Lymphocytes Induce Neuronal. Cell Death in a Human ipsc-based. Model of Parkinson's Disease

Q1 Do you think creating embryos by cell nuclear replacement (CNR) into animal eggs will be beneficial to research?

Gastric Carcinoma with Lymphoid Stroma: Association with Epstein Virus Genome demonstrated by PCR

Morphology and Regional Distribution of the Primordial Germ Cells in the Giant Freshwater Prawn, Macrobrachium rosenbergii

DRB666 Applied Developmental and Reproductive Biology Spring Semester, 2011

BHP 2-7 and Nthy-ori 3-1 cells were grown in RPMI1640 medium (Hyclone) supplemented with 10% fetal bovine serum (Gibco), 2mM L-glutamine, and 100 U/mL

CHAPTER 4 RESULTS. showed that all three replicates had similar growth trends (Figure 4.1) (p<0.05; p=0.0000)

DRB666 Applied Developmental and Reproductive Biology (Spring 2013)

Sex Determination and Gonadal Sex Differentiation in Fish

Sexual plasticity of ovarian germ cells in rainbow trout

Online Data Supplement. Anti-aging Gene Klotho Enhances Glucose-induced Insulin Secretion by Upregulating Plasma Membrane Retention of TRPV2

PHYSIOLOGY AND REPRODUCTION. Fertility of Male and Female Broiler Breeders Following Exposure to Elevated Ambient Temperatures 1

Supplementary information

M.Sc. (Reproductive Biology & Clinical Embryology)

SUPPORTING INFORMATION

Basic information on the cryopreservation process

Beta Thalassemia Sami Khuri Department of Computer Science San José State University Spring 2015

Transcription:

Evidence of in vivo transfection of circulating primordial germ cells in chickens M. NAITO 1 *, T. HARUMI 2, Y. MATSUBARA 1 and T. KUWANA 3 1 Transgenic Animal Research Center, 2 Animal Genome Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki 305-8602, Japan; 3 Laboratory of Intellectual Fundamentals for Environmental Studies, National Institute for Environmental Studies, Tsukuba, Ibaraki 305-8506, Japan. *Corresponding author: e-mail: mnaito@affrc.go.jp Primordial germ cells (PGCs) are progenitor cells of ova and spermatozoa. They appear in the centre of the area pellucida at stage X, and circulate in the bloodstream before migrating to the germinal ridges. In order to introduce exogenous DNA into the germline of chickens, in vivo transfection of circulating PGCs was attempted. DNA-liposome complex was injected into the bloodstream of embryos at stages 14-15, and the manipulated embryos were cultured up to 20.5 days of incubation. GFP gene expression was analysed in the gonads of developing embryos. When circular plasmid was introduced, GFP gene was efficiently expressed in the gonads of embryos, especially up to 10.5 days of incubation. Thereafter, the GFP gene expression decreased gradually, but GFP gene expression was still observed in the gonads of embryos at 20.5 days of incubation. When linearised plasmid was introduced, GFP gene expression was not observed in the gonads of embryos at 20.5 days of incubation except for two (0.45%) embryos. In these two embryos, strong GFP gene expression was observed in a limited area of the left ovary and the left testis. PCR analysis of the gonads of manipulated embryos showed that GFP gene introduced into the PGCs was gradually lost, and most of it by the hatching stage. Nevertheless, GFP gene was still detectable at a very low frequency in some of the gonads of embryos at 20.5 days of incubation. These results suggest that it is possible to introduce exogenous DNA into gonadal germ cells by transfecting circulating PGCs in vivo. Keywords: GFP gene; gonadal germ cell; gonad; primordial germ cell; transfection Introduction The production of transgenic chickens provides numerous applications in the field of animal biotechnology such as producing pharmaceutical materials in eggs as well as studies on the function of cloned genes. The introduction of exogenous DNA into chickens still poses difficulties mainly due to their unique reproductive characteristics and the yolk-laden structure of the ovum (Naito, 2003ab; Sang, 2004). For practical applications of avian transgenic technology, the non-viral method is preferable for safety reasons. The only non-viral method that has successfully produced transgenic chickens to date is microinjection of DNA into the germinal disc of the fertilised ovum (Love et al., 1994; Sherman et al., 1998). By this method, however, only a limited number of ova can be manipulated because only one ovum can be obtained from each hen. Primordial germ cells (PGCs) are the progenitor cells of ova and spermatozoa. PGCs in chickens are mainly located in the central disc of the area pellucida at stage X (Eyal-Giladi and Kochav, 1976; Ginsburg, 1994; Naito et al., 2001a), and move to the germinal crescent region at stage 4 (Hamburger and Hamilton, 1951). They then enter into the developing blood vessels and circulate temporarily throughout the circulatory system, finally migrating to the germinal ridges, i.e., the future gonads (Kuwana, 1993; Naito, 2003c). The introduction of exogenous DNA into PGCs has been attempted at various developmental stages, e.g., PGCs or their precursors isolated from stage X blastoderm

(Carsience et al., 1993), PGCs isolated from embryonic blood (Naito et al., 1998), or PGCs isolated from embryonic gonads (Hong et al., 1998) have been transfected in vitro and then transferred to the recipient embryos. The introduced DNA was expressed in those embryos, but most of it was present episomally and disappeared during embryonic development. The integration of exogenous DNA into the host chromosomes of PGCs is expected to occur in a small proportion of the in vitro manipulated PGC population, but so far no PGCs have been obtained in which exogenous DNA was integrated into the chromosomes, probably due to the limited number of PGCs that could be manipulated in vitro. Although the culture of PGCs in vitro has been successful, the proliferation rate is still very low (Naito et al., 2001b; Han et al., 2002; Park et al., 2003). In vivo manipulation of PGCs has the advantage of introducing exogenous DNA into germ cells. When PGCs are transfected in vivo, all PGCs in the embryo can be manipulated and are able to proliferate rapidly after entering the gonads. Watanabe et al. (1994) attempted to transfect PGCs in vivo by lipofection. In their experiment, liposome-dna complexes were injected into the bloodstream of 2.5-day incubated embryos. The introduced DNA (lacz gene) was expressed in the gonads of developing embryos after 2-3 days of incubation following the manipulation, and the number of laczpositive PGCs was 0.2-2.1 per embryo. Although the transfection efficiency of PGCs in vivo was thus very low, the introduction of exogenous DNA into PGCs in vivo by lipofection was shown to be possible. Recent developments in lipofection technology, however, are expected to enhance the in vivo transfection efficiency of PGCs circulating in the bloodstream. The present study was performed to improve the transfection efficiency of the above technique using green fluorescent protein (GFP) gene as a marker, enabling us to achieve a highly efficient transfection of PGCs in vivo. Materials and methods Fertilised eggs and animal care Fertilised eggs of Barred Plymouth Rock chickens were obtained by artificial insemination from the genetic stock maintained at the National Institute of Livestock and Grassland Science. All animals received humane care as outlined in the Guide for the Care and Use of Experimental Animals (National Institute of Agrobiological Sciences, Animal Care Committee). Transfection of PGCs in vivo and embryo culture Transfection of PGCs was carried out by lipofection using cationic lipids (LA2000). Five microliters of LA2000 solution was first diluted with Opti-MEM I and incubated for 5 min at room temperature (25 ). Two micrograms of plasmid DNA (pbaegfp; GFP gene under the control of chicken β-actin gene promoter) diluted with Opti-MEM I was added, mixed gently, and incubated for 20 min at room temperature. Freshly laid and unincubated fertilised eggs (stage X) were broken, and the contents put in glass vessels. The thick albumen capsule was removed from the yolk, and the embryo (yolk) was transferred to a small host eggshell, filled with thin albumen, covered with cling film, and secured with plastic rings and elastic bands (Perry, 1988; Naito et al., 1990). The reconstituted eggs were cultured at 38, relative humidity 50-60%, for about 53 hrs in a forced-air incubator. When the embryos reached to stages 13-14, the prepared transfection medium (DNA-liposome complexes) was injected into the dorsal aorta of each embryo at a volume of 0.5 μl (50 ng DNA). The control medium was also injected into the bloodstream in the same manner. The manipulated embryos were then transferred to large host eggshells and incubated for up to 20.5 days (Perry, 1988; Naito et al., 1990). Detection of GFP gene expression in gonads of embryos Manipulated and cultured embryos were removed from the yolk, washed with PBS(-). The gonads were then exposed, and the GFP gene expression was observed under a fluorescent microscope (MZFL-III, Leica Microsystems). Detection of GFP gene in gonads of embryos Gonads were removed from the embryos which developed to 20.5 days of incubation and washed with PBS(-). DNA was extracted from the gonads using a DNA extraction kit (SepaGene, Sanko Junyaku) according to the manufacturer s instructions. The extracted DNA was dissolved in TE buffer.

PCR analysis was carried out to detect the presence of the GFP gene. The PCR reaction was performed using a programmable thermal controller (Model 9700, Perkin Elmer). The primers for detecting the GFP gene were as follows; 5 -TTC AAG TCC GCC ATG CCC GAA-3, 5 -ATG GGG GTG TTC TGC TGG TAG-3 (Sano et al., 2003). After an initial denaturation step at 94 for 5 min, 40 cycles of amplification were performed; DNA was denatured at 94 for 30 sec, annealed at 60 for 30 sec, and extended at 72 for 30 sec. The reactions were then incubated at 72 for 5 min. For internal control, a sequence of the endogenous glyceraldehydes-3-phosphate dehydrogenase (GAPDH) gene was amplified using the following primers; 5 -CAG ATC AGT TTC TAT CAG C-3, 5 -TGT GAC TTC AAT GGT GAC A-3 (Love et al., 1994). After an initial denaturation step at 94 for 5 min, 40 cycles of amplification were performed; DNA was denatured at 94 for 30 sec, annealed at 55 for 30 sec, and extended at 72 for 30 sec. The reactions were then incubated at 72 for 5 min. The PCR products of each reaction were separated on a 2% agarose gel, and the bands were visualised under UV light after ethidium bromide staining. Results Viabilities of the manipulated embryos Viabilities of the manipulated embryos at 20.5-days of incubation which were injected with DNA-liposome complexes were shown in Table 1. When circular form DNA was used for lipofection, viability of the embryos was 44.6% (183/410), whereas when linearised DNA was used, viability was 69.7% (442/634). Expression of the GFP gene in gonads of developing embryos GFP gene expression in the gonads was observed only when circular form plasmid DNA was transfected; that expression in the gonads of manipulated embryos was shown in Figure 1. A strong GFP gene expression was detected in the gonads of embryos up to 10.5 days of incubation. During this period, that strong GFP gene expression was observed throughout the gonads. After that, GFP gene expression gradually decreased as the incubation continued, but it was still detected in the gonads of 20.5-day incubated embryos just before hatching, although it was only faintly expressed. Green fluorescence was not observed at any developmental stage in the embryos injected with the control medium (lacking LF2000 or plasmid DNA). Expression and presence of GFP gene in gonads of 20.5-day incubated embryos The manipulated embryos that developed to 20.5 days of incubation were examined for the expression and presence of the GFP gene in the gonads. That expression was observed in 86.3% (158/183) of embryos examined only when circular form plasmid DNA was transfected (Table 1), whereas its presence in the gonads was detected in only one embryo (0.55%, 1/158) when the circular form was transfected (Table 1). The GFP gene was detected in the gonads of 4.30% (19/442) of embryos analysed when linearised plasmid DNA was transfected. Figure 1. Expression of GFP gene in the gonads of developing chicken embryos at 6.5 days (A) and 7.5 days (B) of incubation. Circulating primordial germ cells (st. 14) were transfected in vivo by lipofection at 2.5 days of incubation, and the manipulated embryos were cultured in host eggshells. GFP gene expression was observed throughout the gonads.

Table 1. Presence and expression of GFP gene in gonads of 20.5 day-incubated embryos transfected in vivo by lipofection at 2.5 days of incubation Plasmid (pbaegfp) Number of embryos treated Number (%) of embryos surviving at 20.5 days of incubation Number (%) of embryos expressing GFP gene in gonads of 20.5 days of incubation Number (%) of GFPpositive gonads by PCR at 20.5 days of incubation Circular 410 183 (44.6) 158 (86.3) 1 (0.55) Linear 634 442 (69.7) 2 (0.45) 19 (4.30) Discussion Avian species show some unique characteristics of PGC migration such as circulating in the bloodstream before colonising the gonads. The present study demonstrates that circulating PGCs can be efficiently transfected in vivo by lipofection and, as a result, the introduced GFP gene was strongly expressed throughout the gonads when circular form plasmid DNA was used for transfection. This strong GFP gene expression persisted up to 10.5 days of incubation, i.e., 8 days after the manipulation. Thereafter, GFP gene expression decreased gradually, but was still detectable at 20.5 days of incubation just before hatching. At this stage, GFP was observed in 86.3% of embryos examined, but the presence of the GFP gene in the gonads was detected in only one (0.55%). These results suggest that the GFP gene was efficiently introduced into the PGCs circulating in the bloodstream by in vivo lipofection, and that the transfected PGCs colonised in the gonads and strongly expressed the GFP gene. Although this strong GFP gene expression persisted during the first 8 days after manipulation, the GFP gene was gradually lost, and most of it had disappeared by the hatching stage. The number of PGCs circulating in the bloodstream reaches a maximum at stages 14-15 and starts to migrate to the germinal ridges at stages 15-16, although there are egg-to-egg variations in the number of PGCs present at the same developmental stage (Kuwana, 1993; Tajima et al., 1999; Zhao et al., 2003). Transfection of PGCs circulating in the bloodstream in vivo can, therefore, be carried out by injecting DNA-liposome complexes into the bloodstream at stage 14, as suggested by Watanabe et al. (1994). Since DNA-liposome complexes are stable for several hrs even in the presence of serum, most of the PGCs were transfected efficiently during their circulation in the bloodstream, therefore, a strong GFP gene expression throughout the gonads could be achieved by manipulating embryos at the appropriate developmental stage. PGCs proliferate rapidly after entering the gonads and, during such a rapid proliferation phase, exogenous DNA (GFP gene) would have a chance to enter the chromosomes of PGCs or germ cells. When the linearised form of plasmid DNA was used for transfection, GFP gene was detected in the gonads of 4.3% of embryos examined at 20.5 days of incubation. These results suggest that the integration of the GFP gene into the chromosomes of PGCs or germ cells could be expected using the current in vivo lipofection procedure, although the frequency is very low. The viability of embryos at 20.5 days of incubation was higher when the linearised form of plasmid DNA was used for transfection of PGCs compared with that when the circular form was used. Excessive expression of the GFP gene seems to be harmful to developing embryos, although such GFP gene expression was mainly observed in the gonads. The analysis of same-sex and mixed-sex germline chimaeric chickens has shown that PGCs differentiate to ova in the ovary and to spermatozoa in the testes irrespective of their genetic sex (Kagami et al., 1995, 1997; Naito et al., 1999, 2001a). Many genes involved in the sexual differentiation of embryos have been identified, and these genes express transiently at the appropriate period of embryonic development (Shimada, 2002; Smith and Sinclair, 2004). Although genes expressing in PGCs at the stage of sexual differentiation are largely unknown, exogenous genes can be expressed strongly and transiently using the present procedure of in vivo lipofection. The present procedure will, therefore, be useful in studying the functions of genes involved in the differentiation of PGCs.

Acknowledgements The authors would like to thank the staff of the Poultry Management Section of the National Institute of Livestock and Grassland Science for taking care of the birds and providing the fertilised eggs. This study was supported by the Special Coordination Fund (to MN and TK) from the Ministry of Education, Culture, Sports, Science and Technology of the Japanese Government and a Grant-in- Aid (No. 16380193 to MN) from the Japan Society for the Promotion of Science. References Carsience RS, Clark ME, Verrinder Gibbins AM, Etches RJ. 1993. Germline chimeric chickens from dispersed donor blastodermal cells and compromised recipient embryos. Development 117:669-675. Eyal-Giladi H, Kochav S. 1976. From cleavage to primitive streak formation: A complementary normal table and a new look at the first stages of the development of the chick I. General morphology. Developmental Biology 49:321-337. Ginsburg M. 1994. Primordial germ cell formation in birds. In: Marsh J, Goode J, editors. Ciba Foundation Symposium 182, Germline Development. Chichester, UK: John Wiley and Sons, p.52-67. Hamburger V, Hamilton HL. 1951. A series of normal stages in the development of the chick embryo. Journal of Morphology 8:49-92. Han JY, Park TS, Hong YH, Jeong DK, Kim JN, Kim KD, Lim JM. 2002. Production of germline chimeras by transfer of chicken gonadal primordial germ cells maintained in vitro for an extended period. Theriogenology 58:1531-1539. Hong YH, Moon YK, Jeong DK, Han JY. 1998. Improved transfection efficiency of chicken gonadal primordial germ cells for the production of transgenic poultry. Transgenic Research 7:247-252. Kagami H, Clark ME, Verrinder Gibbins AM, Etches RJ. 1995. Sexual differentiation of chimeric chickens containing ZZ and ZW cells in the germ-line. Molecular Reproduction and Development 42:379-387. Kagami H, Tagami T, Matsubara Y, Harumi T, Hanada H, Maruyama K, Sakurai M, Kuwana T, Naito M. 1997. The developmental origin of primordial germ cells and the transmission of the donor-derived gametes in mixed-sex germline chimeras to the offspring in the chicken. Molecular Reproduction and Development 48:501-510. Kuwana T. 1993. Migration of avian primordial germ cells toward the gonadal anlage. Development Growth and Differentiation 35:237-243. Love J, Gribbin C, Mather C, Sang H. 1994. Transgenic birds by DNA microinjection. Bio/technology 12:60-63. Naito M, Nirasawa K, Oishi T. 1990. Development in culture of the chick embryo from fertilized ovum to hatching. Journal of Experimental Zoology 254:322-326. Naito M, Sakurai M, Kuwana T. 1998. 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 113:137-143. Naito M, Matsubara Y, Harumi T, Tagami T, Kagami H, Sakurai M, Kuwana T. 1999. Differentiation of donor primordial germ cells into functional gametes in the gonads of mixed-sex germline chimaeric chickens produced by transfer of primordial germ cells isolated from embryonic blood. Journal of Reproduction and Fertility 117:291-298. Naito M, Sano A, Matsubara Y, Harumi T, Tagami T, Sakurai M, Kuwana T. 2001a. Localization of primordial germ cells or their precursors in stage X blastoderm of chickens and their ability to differentiate into functional gametes in opposite-sex recipient gonads. Reproduction 121:547-552. Naito M, Sano A, Tagami T, Harumi T, Matsubara Y, Kuwana T. 2001b. Effect of mytomycin C treatment on feeder cells and the proliferation of cultured primordial germ cells isolated from embryonic blood. Journal of Poultry Science 38:343-347.

Naito M. 2003a. Development of avian embryo manipulation techniques and their application to germ cell manipulation. Animal Science Journal 74:157-168. Naito M. 2003b. Genetic manipulation in chickens. World s Poultry Science Journal 59:361-371. Naito M. 2003c. Cryopreservation of avian germline cells and subsequent production of viable offspring. Journal of Poultry Science 40:1-12. Park TS, Jeong DK, Kim JN, Song GH, Hong YH, Lim JM, Han JY. 2003. Improved germline transmission in chicken chimeras produced by transplantation of gonadal primordial germ cells into recipient embryos. Biology of Reproduction 68:1657-1662. Perry MM. 1988. A complete culture system for the chick embryo. Nature 331:70-72. Rapp JC, Harvey AJ, Speksnijder GL, Hu W, Ivarie R. 2003. Biologically active human interferon α- 2b produced in the egg white of transgenic hens. Transgenic Research 12:569-575. Sang H. 2004. Prospects for transfenesis in the chick. Mechanisms of Development 121:1179-1186. Sano A, Tagami T, Harumi T, Matsubara Y, Naito M. 2003. Introduction of exogenous DNA into gonads of chicken embryos by lipofection and electroporation of stage X blastoderms in vivo. British Poultry Science 44:36-39. Sherman A, Dawson A, Mather C, Gilhooley H, Li Y, Mitchell R, Finnegan D, Sang H. 1998. Transposition of the Drosophila element mariner into the chicken germ line. Nature Biotechnology 16:1050-1053. Shimada K. 2002. Sex determination and sex differentiation. Avian and Poultry Biology Reviews 13:1-14. Smith C, Sinclair A. 2004. Sex determination: insights from the chicken. BioEssays 26:120-132. Tajima A, Hayashi H, Kamizumi A, Ogura J, Kuwana T, Chikamune T. 1999. Study on the concentration of circulating primordial germ cells (cpgcs) in early chick embryos. Journal of Experimental Zoology 284:759-764. Watanabe M, Naito M, Sasaki E, Sakurai M, Kuwana T, Oishi T. 1994. Liposome-mediated DNA transfer into chicken primordial germ cells in vivo. Molecular Reproduction and Development 38:268-274. Zhao D-F, Yamashita H, Matsuzaki M, Takano T, Abe S-I, Naito M, Kuwana T. 2003. Genetic factors affect the number of circulating primordial germ cells in early chick embryos. Journal of Poultry Science 40:101-113.