Saturday, January 29, 2011

How Much Would A Dark Weave Cost

(MALE AND FEMALE) OF BOTH PARENTS

, Deng1 Jian Min, 2 *, Kei Satoh1, 3 *, Hao Chang1, 2.4, Zhaoping Zhang1, M. David Stewart1, Hongren
Wang5, Austin J. Cooney5, and Richard R. Behringer1, 2,3,4 **
1Department of Genetics and 2Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA, 3Department of Biochemistry and Cell Biology, Rice University, Houston
TX 77251, USA; 4 Programme in Genes and Development, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA; 5Department of Molecular and Cellular Biology, Baylor College of
Medicine, Houston, TX 77030, USA
* Both authors contributed equally
Short Title: Generation of viable mice from two parents
Summary Judgement: XO cells pluripotent stem cells derived from XY cells derived somatic differentiated to functional oocytes in women's fantasies, which can be fertilized to produce a normal male and female offspring with alleles taken from two parents.


Grant Support: Supported by grants from the National Institutes of Health (NIH) GM81627 AJC and RRB, and Ben F. Love seat, and the Kleberg Foundation to RRB veterinary animal care was supported by Grant Support Cancer Center NIH CA16672.
** Correspondence and reprint request: Department of Genetics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, USA. Tel: 713-834-6427; Fax: 713-834-6339, Email: rrb@mdanderson.org


Summary
In sexual species, fertilization of oocytes produced out individuals with alleles from both parents. Here we use pluripotent stem cells from somatic cells taken from combining the haploid genomes of two males to produce viable sons and daughters. Cells of male mice (XY) were induced pluripotent stem cells (iPS) (father * 1) were used to isolate sub-populations that had spontaneously lost the Y chromosome to be genetically female (XO). These XO males removed were used to generate female chimeras were bred with males genetically different (parent * 2),
Progeny has the genetic information was taken from both parents.
Thus, functional oocytes can be generated from somatic male cells after a new investment program spontaneous sex. These new findings have implications for reproduction in mammals and reproductive assistance technology. INTRODUCTION


natural couplings between males and females cause the fertilization of oocytes by sperm and the generation of progeny that inherited alleles from both parents. In mammals, embryos generated experimentally produced only oogenesis alleles or only spermatogenesis usually die during development due to imbalances in imprinting.Así gene expression, epigenetic differences scheduled for male and female gametogenesis are required for normal mouse development. Interestingly, viable mice "bi-maternal" women have been generated by nuclear transfer into oocytes [4]. This required the introduction of H19 mutant nuclei oocytes were not in the wild type oocytes crops. The frequency of viable mice from these oocytes reconstructed with genetic information taken from two, is less than 1%.





















BIBLIOGRAFIA

1. McGrath J, Solter Solter D. Completion of mouse embryogenesis requires both the maternal and
paternal genomes. Cell 1984; 37:179-183.
2. Surani MA, Barton SC, Norris ML. Development of reconstituted mouse eggs suggests
imprinting of the genome during gametogenesis. Nature 1984; 308:548-550.
3. Mann JR, Lovell-Badge RH. Inviability of parthenogenones is determined by pronuclei, not
egg cytoplasm. Nature 1984; 310:66-67.
4. Kono T, Obata Y, Wu Q, Niwa K, Ono Y, Yamamoto Y, Park ES, Seo JS, Ogawa H. Birth of
parthenogenetic mice that can develop to adulthood. Nature 2004; 428:860-864.
5. Kawahara M, Wu Q, Takahashi N, Morita S, Yamada K, Ito M, Ferguson-Smith AC, Kono T.
High-frequency generation of viable mice from engineered bi-maternal embryos. Nat Biotechnol
2007; 25:1045-1050.
6. Kawahara M, Kono T. Longevity in mice without a father. Hum Reprod 2010; 25:457-461.
7. Jäger RJ, Anvret M, Hall K, Scherer G. A human XY female with a frame shift mutation in
the candidate testis-determining gene SRY. Nature 1990; 348:452-454.
8. Lovell-Badge R, Robertson E. XY female mice resulting from a heritable mutation in the
primary testis-determining gene, Tdy. Development 1990; 109:635-646.
9. Dumic M, Lin-Su K, Leibel NI, Ciglar S, Vinci G, Lasan R, Nimkarn S, Wilson JD,
McElreavey K, New MI. Report of fertility in woman with predominantly 46,XY karyotype in
family with multiple disorders of sexual development: review of prismatic case. Mt Sinai J Med
2008; 75:168-169.
10. Eicher EM, Washburn LL, Whitney JB 3rd, Morrow KE. Mus poschiavinus Y chromosome
in the C57BL/6J murine genome causes sex reversal.
Science 1982; 217:535-537.
Deng 8
11. Nagamine CM, Taketo T, Koo GC. Studies on the genetics of tda-1 XY sex reversal in the
mouse. Differentiation 1987; 33:223-231.
12. Park EH, Taketo T. Onset and progress of meiotic prophase in the oocytes in the B6.YTIR
sex-reversed mouse ovary. Biol Reprod 2003; 69:1879-1889.
13. Otsuka S, Konno A, Hashimoto Y, Sasaki N, Endoh D, Kon Y. Oocytes in newborn MRL
mouse testes. Biol Reprod 2008; 79:9-16.
14. Bradley A, Evans M, Kaufman MH, Robertson E. Formation of germ-line chimaeras from
embryo-derived teratocarcinoma cell lines. Nature 1984; 309:255-256.
15. Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying QL, Smith A.
Capture of authentic embryonic stem cells from rat blastocysts. Cell 2008; 135:1287-1298.
16. Li P, Tong C, Mehrian-Shai R, Jia L, Wu N, Yan Y, Maxson RE, Schulze EN, Song H,
Hsieh CL, Pera MF, Ying QL. Germline competent embryonic stem cells derived from rat
blastocysts. Cell 2008; 135:1299-1310.
17. Feng B, Ng JH, Heng JC, Ng HH. Molecules that promote or enhance reprogramming of
somatic cells to induced pluripotent stem cells. Cell Stem Cell 2009; 4:301-312.
18. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent
stem cells. Nature 2007; 448:313-317.
19. Eggan K, Rode A, Jentsch I, Samuel C, Hennek T, Tintrup H, Zevnik B, Erwin J, Loring J,
Jackson-Grusby L, Speicher MR, Kuehn R, Jaenisch R. Male and female mice derived from the
same embryonic stem cell clone by tetraploid embryo complementation. Nat Biotechnol 2002;
20:455-459.
20. Kobayashi A, Shawlot W, Kania A, Behringer RR. Requirement of Lim1 for female
reproductive tract development. Development 2004; 131:539-549.
21. Sybert VP, McCauley E. Turner's syndrome. N Engl J Med 2004; 351:1227-1238.
22. Russell WL, Russell LB, Gower JS. Exceptional inheritance of a sex-linked gene in the
mouse explained on the basis that the X/O sex-chromosome constitution is female. Proc Natl
Acad Sci USA 1959; 45:554–560.
23. Cattanach BM. XO mice. Genet Res 1962; 3:487-490.
24. Szabó PE, Hübner K, Schöler H, Mann JR. Allele-specific expression of imprinted genes in
mouse migratory primordial germ cells. Mech Dev 2002; 115:157-160.
25. Nagy A, Gertsenstein M, Vintersten K, Behringer R. Manipulating the mouse Embryo: A
Laboratory Manual, 3rd ed. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press;
2003.
26. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and
adult fibroblast cultures by defined factors. Cell 2006; 126:663-676.
27. Meissner A, Wernig M, Jaenisch R. Direct reprogramming of genetically unmodified
fibroblasts into pluripotent stem cells. Nat Biotechnol 2007; 25:1177-1181.
28. O'Gorman S, Dagenais NA, Qian M, Marchuk Y. Protamine-Cre recombinase transgenes
efficiently recombine target sequences in the male germ line of mice, but not in embryonic stem
cells. Proc Natl Acad Sci USA 1997; 94:14602-14607.
29. Bishop CE, Hatat D. Molecular cloning and sequence analysis of a mouse Y chromosome
RNA transcript expressed in the testis. Nucleic Acids Res 1987; 15:2959-2969.
30. Uchida M, Tokunaga T, Aizawa S, Niwa K, Imai H. Isolation and nature of mouse
embryonic stem cell line efficiently producing female germ-line chimeras. Anim Sci Technol
(Jpn) 1995; 66:361-367.
31. Tomizuka K, Yoshida H, Uejima H, Kugoh H, Sato K, Ohguma A, Hayasaka M, Hanaoka
Deng 9
K, Oshimura M, Ishida I. Functional expression and germline transmission of a human
chromosome fragment in chimaeric mice. Nat Genet 1997; 16:133-143.
32. Sato H, Amagai K, Shimizukawa R, Tamai Y. Stable generation of serum- and feeder-free
embryonic stem cell-derived mice with full germline-competency by using a GSK3 specific
inhibitor. Genesis 2009; 47:414-422.
33. Gubbay J, Collignon J, Koopman P, Capel B, Economou A, Münsterberg A, Vivian N,
Goodfellow P, Lovell-Badge R. A gene mapping to the sex-determining region of the mouse Y
chromosome is a member of a novel family of embryonically expressed genes. Nature 1990;
346:245-250.
34. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S.
Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;
131:861-872.
35. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J,
Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem cell lines
derived from human somatic cells. Science 2007; 318:1917-1920.
36. Stadtfeld M, Hochedlinger K. Induced pluripotency: history, mechanisms, and applications.
Genes Dev 2010; 24:2239-2263.
37. Urbach A, Benvenisty N. Studying early lethality of 45,XO (Turner's syndrome) embryos
using human embryonic stem cells. PLoS One 2009; 4:e4175.
38. Zhou GB, Meng QG, Li N. In vitro derivation of germ cells from embryonic stem cells in
mammals. Mol Reprod Dev 2010; 77:586-594.
39. Lee J, Kanatsu-Shinohara M, Ogonuki N, Miki H, Inoue K, Morimoto T, Morimoto H,
Ogura A, Shinohara T. Heritable imprinting defect caused by epigenetic abnormalities in mouse
spermatogonial stem cells. Biol Reprod 2009; 80:518-527.
40. Hübner K, Fuhrmann G, Christenson LK, Kehler J, Reinbold R, De La Fuente R, Wood J,
Strauss JF 3rd, Boiani M, Schöler HR. Derivation of oocytes from mouse embryonic stem cells.
Science 2003; 300:1251-1256.
41. Nicholas CR, Haston KM, Grewall AK, Longacre TA, Reijo Pera RR. Transplantation
directs oocyte maturation from embryonic stem cells and provides a therapeutic strategy for
female infertility. Hum Mol Genet 2009; 18:4376-4389.
42. Behringer RR. Human-animal chimeras in biomedical research. Cell Stem Cell 2007; 1:259-
262.
43. Meaburn KJ, Parris CN, Bridger JM. The manipulation of chromosomes by mankind: the
uses of microcell-mediated chromosome transfer. Chromosoma 2005; 114:263-274.
44. O'Doherty A, Ruf S, Mulligan C, Hildreth V, Errington ML, Cooke S, Sesay A, Modino S,
Vanes L, Hernandez D, Linehan JM, Sharpe PT, Brandner S, Bliss TV, Henderson DJ, Nizetic
D, Tybulewicz VL, Fisher EM. An aneuploid mouse strain carrying human chromosome 21 with
Down syndrome phenotypes. Science 2005; 309:2033-2037.