干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 255410|回复: 245
go

Variable Reprogramming of the Pluripotent Stem Cell Marker Oct4 in Mouse Clones: [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:49 |只看该作者 |倒序浏览 |打印
a Germline Development Group, Center for Animal Transgenesis and Germ Cell Research, New Bolton Center, University of Pennsylvania, Kennett Square, Pennsylvania, USA;
4 A4 j' i( a8 ~0 P: T
" C8 G2 F, K6 R; g- V0 X% pb Laboratory of Developmental Biology, Department of Animal Biology, University of Pavia, Pavia, Italy( ^6 b8 ^+ ?) I$ Z5 O

8 C0 o% l2 i1 y" zKey Words. Embryo ? Embryonic stem cell ? Nuclear transfer ? Oct4 ? Reprogramming
$ t3 t6 X0 }2 s0 d$ `. t% w$ o( J& S( |
Correspondence: Michele Boiani, Ph.D., Max Planck Institute for Molecular Biomedicine, Mendelstra?e 7, D-48149 Münster, Germany. Telephone: 0049-0251-980-2864; Fax: 0049-0251-980-2992; e-mail: mboiani@mpi-muenster.mpg.de! f6 J& G& h) I6 _! }; h, _
+ [0 L  v% a8 `9 o) u
ABSTRACT6 H2 }5 @  o2 Z3 F" F  j5 Z+ u4 o, I
+ \* j) A, b& @# |7 {3 A' {* l
Nuclear transfer techniques are used to study fundamental biologic issues, such as reversibility of differentiated cellular states and interactive processes between the nucleus and the cytoplasm. These aspects are collectively dubbed reprogramming〞a fashionable term in cloning research to indicate that the cellular state of a nucleus changes from that of a differentiated somatic cell into a pluripotent embryonic cell after the nucleus is transplanted into an oocyte. Cellular states are largely determined by the coordinated expression of thousands of genes. Epigenetic mechanisms (including DNA methylation and demethylation, post-translational histone modifications, and swapping of histone variants as well as other chromatin-associated proteins) are thought to underlie the somatic-to-embryonic transition of gene expression in the transplanted nucleus and the maternal-to-embryonic transition of gene expression after fertilization . Although these epigenetic changes always take place to some extent in clonal embryos, they are not always consequential in terms of gene expression . Therefore, reprogramming at the DNA template level demands phenotypic assessment through studies of gene expression and cellular function in clones.
, A* i, `: o# V; M* F5 B( p0 Q
3 `* C( {. M, ?5 ~, ^/ hThe initial experiments on cloning in amphibia by Briggs, King, and Gurdon demonstrated that the potential of the nucleus to direct normal embryonic development decreases progressively with the differentiation stage of the donor cell . This phenomenon is notably evident in mouse cloning experiments using donor nuclei from somatic cells and pluripotent embryonic stem (ES) cells . After birth, Sertoli cell–cloned mice tend to die prematurely of hepatic failure or tumors  and cumulus cell–cloned mice tend to grow obese , whereas ES cell–cloned mice survive in larger numbers〞provided they originate from F1 nucleus donors . In these cases, the epigenetic make up of the donor nucleus exerts some priming and lasting effects on the clonal phenotype. Similarly, the proportion of fibroblast-derived and cumulus cell–derived bovine clones having zygote-like epigenotypes (as defined by patterns of methylation and acetylation of histone H3 lysine 9) correlates closely with the proportion developing to the blastocyst stage .
, ]! H7 M, v* p; j3 e! l. C
+ q" D" G' _* ?, w7 e8 a% qAlthough a nucleus-driven scenario for clones is logically appealing, environmental effects should be considered as well. In isogenic Avy and Axin(Fu) mice, the coat color and tail phenotypes show extremely variable expressivity linked to the activity of an inserted retrotransposon , and diet can alter the methylation status of the retrotransposon and cause the phenotype change . Differences between embryos of mouse strains that are traditionally considered blocking and nonblocking in vitro at the two-cell stage are contingent on the culture medium osmolarity . In somatic cells, cytoplasmic compartments and pools of molecules are able to reallocate in response to changes in the culture medium composition . In somatic cell–derived clonal embryos, certain idiosyncrasies of myoblast and cumulus cell clones referred to as somatic cell–like features are well documented but their causes remain elusive . Environmental cues seem to merely modulate the developmental competence of already established zygotes , but the developmental competence of somatic cell clones is not yet established at nuclear transfer. In intact mouse oocytes, nuclear remodeling activities continue to operate after activation , albeit not to the extent observed when donor nuclei are exposed to metaphase II ooplasms . Alteration of pronuclear remodeling by culture conditions after oocyte fertilization would lead to a suboptimal, yet still zygotic, genetic program of embryonic development. In contrast, alteration of nuclear remodeling after somatic-cell nuclear transfer may either support or prevent the establishment of a zygotic genetic program, depending on the specific cues from the environment.7 |& O! Z* U. F1 Y: [8 k2 U
# G  {. J% r9 k- S. H) m3 M! K- U
Is the reprogramming of the donor nucleus also contingent on the culture environment of the clone? The focus of this study was to investigate whether developmental pluripotency after cumulus-cell nuclear transfer ensues independently from, or is conditioned by, the culture environment as tested in a variety of forms.# |8 _4 n- V  _7 g; _" _

, u2 g  S6 p, @; E+ e1 k5 a% `+ `( lMATERIALS AND METHODS
8 s8 }6 {- \1 y) }
+ t$ ^  z0 E. R) P+ S3 ]- {& y- kQuality Control for Cumulus Cell Nuclear Transfer4 n4 h2 V, w: f; o

$ D! s# v. n( M+ zIn principle, the differences observed in clones are possible consequences of either manipulation or faulty reprogramming, although Wakayama and colleagues  showed that manipulation and culture are not as detrimental to clonal mouse embryos as often assumed. The fact that cloning results are very variable, while originating from research in a limited number of laboratories, demonstrates the complexity of the technology used and exemplifies the above concern. In this study, possible variations in micromanipulation that might cause differences of development and gene transcription in the clones were neutralized by manipulating the prospective six-media clones using the same setup and only then allocated to the different culture media. We previously showed that oocyte micromanipulation per se does not cause significant reduction of ATP content in clones within 48 hours compared with zygotes . The effect of micro-injection, as measured by development after intracytoplasmic sperm injection (ICSI) into metaphase II oocytes, was negligible in that 87% of the oocytes survived the sperm head injection (n = 1,991/2,301), 91% and 86% of these cleaved to two and four cells, respectively (n = 1714 and 1,816/1,991), 49% formed blastocysts at 96 hours (n = 969/1,991), and 32% of the embryos transferred in vivo at various stages developed to term (n = 40/125). ICSI is routinely performed in our laboratory: data just presented were the average of a 1-year period that encompasses the present study (Boiani, unpublished data). To further assess whether micromanipulation damages the integrity of the transplanted genome, the karyotype of clonal embryos was analyzed (Fig. 1). Karyotype analysis of clonal four-cell stage embryos detected hyperaneuploid sets of chromosomes (>40) in 10.8% ± 3.2% of blastomeres (n = 93). This did not differ significantly (p > .05) from the rate of hyperaneuploidy observed in blastomeres of parthenogenetic (10.1% ± 3.4%; n = 79) and zygotic (8.3% ± 2.4%; n = 133) embryos cultured under the same conditions (Boiani, unpublished data). Gene/allele regulation, rather than physical absence of loci, would therefore seem to be at the root of the different patterns of gene expression expected to be found in clones. Clones and zygotes had an equivalent ooplasmic composition, B6C3, thus preventing strain-specific ooplasmic modifiers from having an unequal effect on the incoming nucleus .
0 o/ e5 C1 i& v1 k3 E" m6 T/ W9 p# }8 A! B& ?4 l' B" v
Culture Conditions Affect the Rate of Blastocyst Formation of Genetically Identical Mouse Clones% P- a& q  A4 }# |, d% n
9 b0 H5 Q5 S, V. }: R
As long as the clones are generated with the same experimental setup and only subsequently allocated to different culture media, as was the case in this study, differences among clones depend on the culture media only. We assessed the developmental program of clonal and zygotic mouse embryos by culturing them in parallel up to the blastocyst stage. To this end, strain B6C3 mouse oocytes were enucleated and transplanted with B6C3 cumulus cell nuclei to derive the clones or were fertilized in vivo with B6C3 sperm to recover one-cell zygotes from the oviducts.$ x. t% \, c4 u

' h9 x8 s2 }5 V1 M+ rAt 96 hours of development in vitro (108 hours after human chorionic gonadotropin), zygotes had formed blastocysts at similarly high rates (80%–85%) in all media tested except DMEM (8%; see Fig. 2A for absolute rates). The rate of clonal blastocyst formation progressively increased (multiple t-tests, p
- q4 v. O- P) y2 m1 E6 m& c: A
0 C( `5 r8 Z/ f( o! \" S! B9 TFigure 2. Rates of blastocyst formation for (A) zygotes and (B) cumulus cell–derived clones cultured side by side in DMEM, G1/G2, CZB, KSOM (aa), M16, and -MEM. Each data point is an average of measurements obtained from 15 independent experiments (number of starting one-cell embryos: 42, 16, 93, 91, 82, 91 zygotes; 246, 90, 490, 418, 415, 443 clones allocated to DMEM, G1/G2, CZB, KSOM(aa), M16, and -MEM, respectively). Error bars indicate the SDs. Abbreviations: DMEM, Dulbecco’s modified Eagle’s medium; MEM, minimal essential medium.
  `- I0 j& Z* P" J
3 v& o. c* Y+ z: |& \; s) tCulture Conditions Modulate Gene Expression in Clones Differently Than in Zygotes( a  c  W: r4 m% {5 ~% L$ z
$ D; u6 e# t: Y
The developmental program that guides the progression of clones through preimplantation stages may result in the formation of either genuine blastocysts or seeming blastocysts that have only the morphological appearance of normal blastocysts (phenocopies ). These phenocopies can be distinguished by their aberrant gene expression that may correlate with subsequent developmental failure, in vivo and also in part in vitro. Although as many as 70% of cumulus cell–derived mouse clones can develop up to the blastocyst stage in vitro, most fail to reproduce the level and spatial pattern of Oct4 gene expression present during zygotic development that confers pluripotency on blastomeres of the morula and the ICM . Therefore, we investigated whether cumulus-cell clones cultured in different media uniformly failed to reactivate the Oct4 gene from the silent somatic state .' g9 m. d/ J1 [6 v
' A# C( y. i/ G6 L6 [: y. k
The expression level of Oct4 and other genes was analyzed in individual blastocysts by real-time Q-RT-PCR as follows: Oct4 (chromosome  17), Bex1 a.k.a. Rex3 (chr X), Gapdh (chr 6), Cpt2 (chr 4), and Hprt (chr X). These genes were chosen for the following reasons: Bex1 is a TE-restricted gene in mouse blasto-cyst development  whereas Oct4 is not , and Gapdh and Cpt2 are metabolic genes that are relevant to the health status of the embryo in culture . Hprt was used for normalization purposes. In theory, differences in the gene expression levels between clonal and zygotic blastocysts might simply reflect differences in the numbers of cells in these two types of blastocysts  or may be due to differences in the state of activity of the genes at the time of nuclear transfer rather than to differential gene regulation due to reprogramming. Hprt was used for normalization purposes after finding it not to be altered significantly in clonal and zygotic blastocysts within the culture medium groups (see ANOVA below). Hprt expression levels are also conserved in male and female zygotic mouse blastocysts ./ C) s% j, W' W- o+ P6 S% r- a4 K

" k7 U  N7 B" z/ ^0 IFive single clonal blastocysts and five single zygotic blastocysts grown in each medium were analyzed by Q-RT-PCR. This approach relies on the buildup of the amplification product with each PCR cycle, which increases SYBR green I light emission until it crosses a threshold; hence, the lower the template amount, the higher the number of cycles. Raw Ct and SDs of clonal and zygotic blastocysts are shown in Table 2. One-way ANOVA of Ct values for Hprt by type of embryo produces nearly identical distributions for clonal and zygotic blastocysts in concert with Hprt as a normalizer (F1,71 = 0.0274, P > F 0.8689). The raw Q-RT-PCR data indicate that Oct4 mRNA amplifies above threshold 4.11 cycles earlier than Hprt in the -MEM medium, whereas it amplifies 3.54 cycles earlier than Hprt in the CZB medium. Thus, there seems to be less Oct4 mRNA in the CZB than in the -MEM sample on an absolute basis, in agreement with the RISH data. However, Q-RT-PCR data presented in raw format are not acceptable as they may merely reflect the amount of total mRNA extracted.
2 G# l  b5 X7 i6 u9 D+ T
7 x4 _. F) y( DTable 2. Amounts of clonal and zygotic blastocyst mRNAs expressed by the crossover threshold (Ct) number, e$ p$ \* z% d  I0 m5 V  n2 X5 M8 A5 w
' R& E( U" G& {2 ^
Therefore, values were normalized to that of Hprt mRNA, and a clone-zygote comparison of Hprt-normalized transcript levels (Table 3) was conducted with the  Ct method (ABI Prism 7.700 User Bulletin 2; PerkinElmer, Wellesley, MA, http://www.perkinelmer.com) . Zygotes assumed a value of 1, and clones that exhibited a higher or lower gene transcript level than zygotes were given a value greater than or less than 1, respectively. Oct4 transcripts were detected in all embryos analyzed, and in general, based on the normalized values, Oct4 was expressed at higher levels in clones than in zygotes (1 to 5.14,  414%). There were also notable variations in Oct4 transcript levels in CZB-cultured clones (1 to 14.96,  1,396%). These results were consistent with the lower Bex1 expression levels (1 to 0.27, –73%) based on opposite regional regulation of Oct4 and Bex1 genes in the blastocyst (Oct4 expression in the ICM, Bex1 in the TE). The level of Cpt2 relative to Hprt mRNA was similar in clones and zygotes across all media (1 to 0.86, –14%), whereas Gapdh mRNA levels fluctuated. Overall, Gapdh exhibited 185% higher (1 to 2.85,  185%) expression in clones compared with zygotes, with 118% of the total increase contributed by mRNA levels in one medium〞CZB. As determined by karyotype and Xist analysis, gene/allele regulation rather than physical gain or loss of chromosomes, or X-chromosome inactivation status, seemed to cause the different patterns of gene expression (see Quality Control for Cumulus Cell Nuclear Transfer and Materials and Methods).- Q4 q( |7 A1 K8 l2 m! u$ J5 j, g

* F* _' X( g9 ?0 _7 ~: uTable 3. Use of the  Ct method (PerkinElmer ABI Prism 7.700 User bulletin n.2) to calculate deviations of clonal versus zygotic blastocysts in transcripts amounts
+ p$ k6 y  f# b0 y( L5 @% V5 I, q6 C: f9 B- \
The expression patterns of the five genes in clones were compared across the different media. This was done by calculating the correlation coefficient (r) between the medium series of gene expression (Table 3) that were paired in all possible ways leaving out reciprocal duplicates (e.g., correlation between Cpt2 and Oct4 expression levels in G1/G2, CZB, KSOM(aa), M16, and -MEM), based on the formula r = covariance /std dev*std dev . Correlation coefficients were as follows: Bex1-Oct4, r = 0.004; Gapdh-Bex1, r = 0.136; Gapdh-Cpt2, r = 0.248; Cpt2-Oct4, r = 0.286; Cpt2-Bex1, r = 0.492; Gapdh-Oct4, r = 0.953 (Gapdh-Gapdh, r = 1). Oct4 and Bex1 patterns (r = 0.004) were actually inversely related (Oct4 and 1/Bex1, r = 0.893). This demonstrated that genes among the set analyzed in clones respond in a variable manner to different culture conditions, particularly Oct4. This seems contradictory to the critical requirement of Oct4 for the establishment of the pluripotent founder cell population in mouse embryos , to the tight Oct4 regulation in mouse ES cells , and to the observation that Oct4 mRNA levels are rather conserved in individual ICM cells of human blastocysts .
, c  x5 N; y4 O$ K- e- }5 M1 ~# @3 b3 M& r
Functional, But Not Spatial, Definition of the Blastocyst ICM Is Altered by Culture Conditions& `! }) E  |7 N* n4 K1 Z0 b/ F
5 I% {' ~6 ?8 K7 P) I9 a
Until recently, quantitative analysis of high-resolution two-dimensional protein gels , mRNA differential display , analysis of expressed sequence tags derived from libraries of various preimplantation stages , analysis of selected genes , and microarray techniques  have been used in gene expression studies. Although these approaches have shed some light on the molecular basis of preimplantation development, they are based on pooled embryos and thus offer limited insight into clonal embryo development wherein embryo-embryo variability occurs, even within the same batch of clones.- ?/ L" G( Y0 ^( X+ p, b- q

8 n$ }2 I5 E3 g" T) L: K7 OAlterations in the transcript levels of genes that are locally/regionally modulated cannot be detected by any mRNA profiling method, Q-RT-PCR, or microarray technology, unless each individual cell is profiled in situ. For instance, higher Oct4 expression levels ( 414% on average; Table 3) and lower Bex1 expression levels (–73% on average) in clones relative to zygotes may indicate an expansion of the ICM compartment relative to a shrunken TE. The fact that all embryos tested positive for Oct4 transcript by Q-RT-PCR where as previous studies (using riboprobes and conventional RT-PCR) found that some tested negative as well  reflects, in part, the different sensitivity of these assays. Furthermore, it suggests that the amount of Oct4 mRNA may be so high but in only a few cells or so minute overall as to be functionally equivalent to zero when assessed spatially in situ.
) L# V5 P" F/ u9 {2 `) B3 x" K. E+ v, c$ \5 I3 k. n
Clonal blastocysts (n = 234 in total from the various media) were analyzed individually for the presence of an ICM, as defined by localized Oct4 mRNA RISH. Differences were observed between CZB, KSOM(aa), M16, and -MEM culture media (DMEM and G1/G2 did not yield enough blastocysts to compensate for losses incurred by RISH). Hybridized blastocysts were categorized  into four groups: having strong Oct4 signal restricted to the ICM as opposed to weak signal; having unrestricted signal (ICM   TE); or lacking signal in both (Fig. 3A). In general, clonal blastocysts had consistently lower rates of strong, ICM-restricted distribution of Oct4 mRNA than zygotic blastocysts cultured in the same medium (p : y. [* f; O5 I; G1 U( v

$ X) F0 {$ i: Z& k. _+ K1 O+ q: J5 w1 N0 TFigure 3. Expression patterns of Oct4 in clonal and zygotic blastocysts detected by RISH. (A): General patterns of Oct4 mRNA distribution in blastocysts. (B): Frequency of pattern A (worst) and D (best) is dependent on the culture medium used for clones. A total of 234 clonal blastocysts (n = 64, 21, 55, 89, 5 in M16, CZB, KSOM(aa), -MEM, and G1/G2, respectively) were examined by RISH. Control blastocysts were obtained in parallel (n = 41, 35, 30, 32, 35) after culture in G1/G2, CZB, KSOM(aa), M16, and -MEM media, respectively. Each data point is an average of measurements from five independent experiments. Error bars indicate SDs. (C): Clonal blastocysts obtained in CZB and -MEM, from the same experiment, documented after simultaneous processing in the RISH reaction and 3 hours of color reaction. Bar = 100 μm. Abbreviations: ICM, inner cell mass; MEM, minimal essential medium; RISH, riboprobe in situ hybridization.
0 X* |  I+ v7 ?) g' L; D4 M8 r" s$ P% p: O5 c: U' N* ~7 G2 U" T
Because Oct4 becomes restricted to the ICM during normal blastocyst formation, differences in the regional distribution of the transcript among clonal and between clonal and zygotic blastocysts may simply reflect differences in cell numbers and allocations to ICM/TE  in these two types of blastocysts rather than differences in gene regulation. Variability in Oct4 mRNA distribution/restriction to the ICM observed in different culture environments was found to coexist with a similar number of embryonic cells allocated to the ICM, that is, 12 ± 5 ICM cells in CZB and 17 ± 7 ICM cells in -MEM (p > .05) (see Materials and Methods). It follows that reinstatement of pluripotency after cumulus cell nuclear transfer is a primary effect of culture conditions on reprogramming rather than a side effect of cell number allocation or faulty chromosome segregation during cleavage of clonal embryos.
/ |' S# J7 D1 c$ S4 b/ Z3 [+ i7 F" C  j3 n
Consequence of the Culture Medium–Dependent Oct4 Gene Reprogramming
& e/ g% j; _- [
! i: O0 d6 }4 c2 UAltered Oct4 levels affect the pluripotency of ES cells , and lack of Oct4 in the ICM negates the further development of mouse blastocysts . Therefore, it seems likely that differences in Oct4 mRNA levels in clonal blastocysts, observed more frequently than in zygotic blastocysts, also have an impact on clonal development and ES cell derivation. To track this, we compared the reprogramming of genetically identical cumulus-cell nuclei in oocytes cultured in different media by measuring Oct4-GFP expression in those nuclei (Fig. 4, see legend for absolute rates). The normal timing of maternal-zygotic transition to Oct4 gene expression occurs in mice at the eight-cell stage . Although the onset of Oct4-GFP expression in zygotes at the eight-cell stage was equally frequent across all media (p > .05), the proportion of eight-cell clones having Oct4-GFP expression at 55 hours was lower in CZB and G1/G2 and higher in -MEM (p
5 B6 s9 _$ b7 e9 w. @7 D0 W# P/ a0 V8 [2 W4 Z* {
Figure 4. Viable monitoring of nuclear reprogramming during clonal embryo cleavage. Onset and frequency of Oct4-GFP expression in eight-cell clonal and zygotic embryos cultured in M16, CZB, KSOM(aa), -MEM, and G1/G2 media; base of data for each bar, 25 embryos. Error bars indicate SDs. Asterisks indicate significant differences in rates of Oct4-GFP expression between zygotes and clones for the particular culture medium used. Abbreviation: MEM, minimal essential medium.
+ \  ]. ?1 u- T. S  h2 u; a% K5 E0 E6 m1 p( b. e; Q5 f  G, f5 `
Figure 5. Stem cell and fetal consequences of clonal embryo epigenetic reprogramming. Culture conditions conducive to clonal blastocyst formation are associated with differences in the potential of the inner cell mass and trophectoderm to give rise to ES cells and fetal implants, respectively. (A): Clonal blastocysts seeded on feeder cells. (A’): Clonal ES cell colony expressing Oct4-GFP at passage 6. (A"): Oct4-GFP clonal ES cells after injection in zygotic blastocysts. (A"’): Germ cell contribution of Oct4-GFP clonal ES cells that were injected into blastocysts to form 13.5 dpc chimeric fetuses. (B): Summary of ES cell and fetal rates of clones formed in CZB and -MEM. Bar = 100 μm. Abbreviations: ES, embryonic stem; MEM, minimal essential medium.) m! E* @" L% \6 \& D/ X- h5 W' X

) r0 O* z; z/ b! q5 q2 YTo this end, we generated and assayed for ES cell potential an additional batch of clones (n = 400) under the worst and best reprogramming conditions for Oct4 (Fig. 3): Half of the clones were cultured in CZB and half in -MEM, in parallel, from the one-cell stage up to the blastocyst stage, followed by derivation of ES cells. These blastocysts were plated in parallel onto feeder cells (Fig. 5A), using a standard method for derivation and culture of ES cells. Cell lines were identified as ES cells by observing the expression of Oct4-GFP after six passages (Fig. 5A’) and by blastocyst injection (see below). The rates of cell line derivation were found to be 8.2% for the CZB-cultured clones (n = 5 cell lines/61 blastocysts) and 39.3% for the -MEM–cultured clones (n = 33 cell lines/84 blastocysts) (p
% o- I# z# C/ o7 N  ?% Q: }; R  F5 Y* {& ?- }4 n
Altogether, these data show that the patterns of Oct4 gene expression in blastocysts cannot only be assessed as done previously  but also arise nonrandomly according to culture medium patterns and that higher rates of blastocyst formation and ES cell potential of clones in vitro did not translate into higher rates of fetal development (Fig. 5B). To determine whether clonal ES cells would be impaired under conditions that impaired fetal development from precursor blastocysts, clonal ES cells from -MEM were injected into the cavity of blastocysts of zygotic embryos and then assessed for germline contribution at 13.5 dpc. Nine out of 13 lines tested contributed to the formation of germ cells (Figs. 5A", 5A"’). Considering ES cells as the equivalent of the ICM, these results suggest that the reprogrammability of the donor nucleus to give rise to an ICM lineage is different from that required to give rise to a TE lineage.
* f7 `- O, E* |0 v: |* P) w; ^1 N, [; F5 Y+ |, a  E1 n
DISCUSSION( o8 G# W" @4 I2 D4 s

2 A! G4 B; }6 U6 K7 K. iThat reprogramming after somatic cell nuclear transfer may indeed occur subsequent to the metaphase II oocyte stage was reported by Eckardt et al.  after this manuscript was submitted.
8 O. @2 ~4 C; Y$ g/ c  Z$ M; _$ h- h( l8 Z& M- _
ACKNOWLEDGMENTS
1 F/ O: _* |( d6 w( j- C5 f' ]2 ~+ I
Santos F, Dean W. Epigenetic reprogramming during early development in mammals. Reproduction 2004;127:643–651.
9 C3 v. Z) d; h* n7 a* A. k6 U, j+ t6 v2 [. A$ S: }+ p7 m
Vickaryous N, Whitelaw E. The role of early embryonic environment on epigenotype and phenotype. Reprod Fertil Dev 2005;17:335–340.
! F4 H  _8 `! N- L: M( z9 @3 y. p/ _0 T' Z3 v5 S5 v
Santos F, Zakhartchenko V, Stojkovic M et al. Epigenetic marking correlates with developmental potential in cloned bovine preimplantation embryos. Curr Biol 2003;13:1116–1121.
$ x5 C: T1 q; ^/ l) P+ q8 N" `' l1 {
/ t( _+ T- O# \4 T* N& lGurdon JB, Byrne JA. The first half-century of nuclear transplantation. Proc Natl Acad Sci U S A 2003;100:8048–8052.
  G; N- I3 F! s- i3 `2 o5 n5 X5 U0 N# s; O3 Y, A# F% j
Eggan K, Akutsu H, Loring J et al. Hybrid vigor, fetal overgrowth, and viability of mice derived by nuclear cloning and tetraploid embryo complementation. Proc Natl Acad Sci U S A 2001;98:6209–6214.1 `0 i. s1 D3 j. `! J1 r

. l$ o& u. m4 z, D6 d/ xRideout WM 3rd, Wakayama T, Wutz A et al. Generation of mice from wild-type and targeted ES cells by nuclear cloning. Nat Genet 2000;24:109–110.
2 [1 H6 I! w9 J& i; q4 E, s, z
* N  F& e  [0 g/ l( a3 HOgonuki N, Inoue K, Yamamoto Y et al. Early death of mice cloned from somatic cells. Nat Genet 2002;30:253–254.+ g- l  j; A8 g- ]6 O. L9 M

9 q8 E$ \0 v  |( a3 |9 [; qTamashiro KL, Wakayama T, Akutsu H et al. Cloned mice have an obese phenotype not transmitted to their offspring. Nat Med 2002;8:262–267.
5 G+ ^- S$ `+ M% d6 p9 k! B. y
9 K" M* N" B- P+ s3 p6 v9 `8 eMorgan HD, Sutherland HG, Martin DI et al. Epigenetic inheritance at the agouti locus in the mouse. Nat Genet 1999;23:314–318.% [; ~! c1 }# S1 T$ }# a: O$ n

6 I9 V, \+ p8 U9 {: R% Z& wRakyan VK, Chong S, Champ ME et al. Transgenerational inheritance of epigenetic states at the murine Axin(Fu) allele occurs after maternal and paternal transmission. Proc Natl Acad Sci U S A 2003;100:2538–2543.
- Y+ K- H# J' D: D8 G$ R- [
' V$ q" f0 ]. P+ O) w# ~: lWaterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol 2003;23:5293–5300.7 [4 s9 o, d7 g' i1 `
/ r1 h+ G( l8 U1 u0 {! l
Hadi T, Hammer MA, Algire C et al. Similar effects of osmolarity, glucose, and phosphate on cleavage past the 2-cell stage in mouse embryos from outbred and F1 hybrid females. Biol Reprod 2005;72:179–187.
4 G2 }0 f. e8 w7 T5 R1 p9 g! ]7 j! J) Y' c/ }* N3 A
Gajewski CD, Yang L, Schon EA et al. New insights into the bioenergetics of mitochondrial disorders using intracellular ATP reporters. Mol Biol Cell 2003;14:3628–3635.
* ~. |3 f4 x0 t" n- p8 @, u
% W& K  J/ l4 F) \( bGao S, Latham KE. Maternal and environmental factors in early cloned embryo development. Cytogenet Genome Res 2004;105:279–284.* P) o: r. o/ G4 [: i0 z+ s, m! w

1 J, ~7 q( r0 E: WHardy K, Spanos S, Becker D et al. From cell death to embryo arrest: mathematical models of human preimplantation embryo development. Proc Natl Acad Sci U S A 2001;98:1655–1660.
$ [( @  V+ {4 |1 U, ]( J: _- ]7 L  [' R. _1 V) r
Szollosi D, Czolowska R, Soltynska MS et al. Remodelling of thymocyte nuclei in activated mouse oocytes: an ultrastructural study. Eur J Cell Biol 1986;42:140–151.
. |/ V# y/ m% T% D2 y9 |+ b% D9 ^+ h3 M( m
Czolowska R, Modlinski JA, Tarkowski AK. Behaviour of thymocyte nuclei in non-activated and activated mouse oocytes. J Cell Sci 1984;69:19–34.
1 C% [4 K6 i/ p; _$ C% X4 t+ S2 S% M: D+ c
Dyban AP, Lee K, O’Neill GT et al. Cytogenetic study of silver-staining NOR in 8-cell-stage mouse blastomeres fused to 1-cell-stage embryos. Development 1988;104:453–463.
6 [7 X% v+ }9 B6 Q. E7 C' |5 y% B. B/ E! B/ y& j  P4 w2 }: J' R
Bordignon V, Clarke HJ, Smith LC. Factors controlling the loss of immunoreactive somatic histone H1 from blastomere nuclei in oocyte cytoplasm: a potential marker of nuclear reprogramming. Dev Biol 2001;233:192–203.
7 H# D- ~! d/ X9 C1 Q# i0 R/ |4 f5 T! o& Q( R! F/ w, w: W
Gao S, Chung YG, Parseghian MH et al. Rapid H1 linker histone transitions following fertilization or somatic cell nuclear transfer: evidence for a uniform developmental program in mice. Dev Biol 2004;266:62–75.
/ L  U8 `6 z, K5 g  {
# P) |" A* Z5 nSzabo PE, Hubner K, Sch?ler H et al. Allele-specific expression of imprinted genes in mouse migratory primordial germ cells. Mech Dev 2002;115:157–160.3 h4 q% V! i- Q
. C3 B! [  o9 b9 I( ^# R
Boiani M, Kehler J, Sch?ler HR. Activity of the germline-specific Oct4-GFP transgene in normal and clone mouse embryos. Methods Mol Biol 2004;254:1–34.  L+ r/ W/ q) d$ w$ e+ b
) Z1 }( i/ G. T5 z! I
Boiani M, Gambles V, Sch?ler H. ATP levels in clone mouse embryos. Cytogenet Genome Res 2004;105:270–278.* D' O6 e+ V' J$ B, h

, I9 c6 a) Z8 g' [7 {Nagy A, Gertsenstein M, Vintersten K et al. Manipulating the mouse embryo, 3rd ed. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press, 2002:164.
3 p7 i7 ~- E. ]( Y/ n& e& N2 @1 }& h6 M, o$ r% @7 S
Chatot CL, Ziomek CA, Bavister BD et al. An improved culture medium supports development of random-bred 1-cell mouse embryos in vitro. J Reprod Fertil 1989;86:679–688.: w- Q( o2 w' j* O2 E% H
- J1 I% J. B& V9 T+ M' l
Barnes FL, Crombie A, Gardner DK et al. Blastocyst development and birth after in-vitro maturation of human primary oocytes, intracytoplasmic sperm injection and assisted hatching. Hum Reprod 1995;10:3243–3247.
  Z. U# g' m' |1 t
1 E' y. ?3 p: S7 O6 @# `: t3 |1 h# g$ XAbbondanzo SJ, Gadi I, Stewart CL. Derivation of embryonic stem cell lines. Methods Enzymol 1993;225:803–823.
3 T. T6 O- `7 h( F' D5 P2 J2 U) P: E' E, v; j3 c* p
Hartshorn C, Rice JE, Wangh LJ. Developmentally-regulated changes of Xist RNA levels in single preimplantation mouse embryos, as revealed by quantitative real-time PCR. Mol Reprod Dev 2002;61:425–436.9 }& o  T1 f/ U/ H- |2 M( m
1 {9 x5 Y6 O* D; k1 J
Hartshorn C, Rice JE, Wangh LJ. Differential pattern of Xist RNA accumulation in single blastomeres isolated from 8-cell stage mouse embryos following laser zona drilling. Mol Reprod Dev 2003;64:41–51.
  S; ~3 t7 b8 J# Q
& `# j( c  C& s* ?9 r! E3 sSteuerwald N, Cohen J, Herrera RJ et al. Analysis of gene expression in single oocytes and embryos by real-time rapid cycle fluorescence monitored RT-PCR. Mol Hum Reprod 1999;5:1034–1039., W, l9 N6 _, |- C/ H2 c" J
( u6 g5 X0 J0 w$ i; v- L
Steuerwald N, Cohen J, Herrera RJ et al. Quantification of mRNA in single oocytes and embryos by real-time rapid cycle fluorescence monitored RT-PCR. Mol Hum Reprod 2000;6:448–453.
6 U$ A" ~* @) e$ `. {: M: p9 X& ?/ R1 W- M
Bortvin A, Eggan K, Skaletsky H et al. Incomplete reactivation of Oct4-related genes in mouse embryos cloned from somatic nuclei. Development 2003;130:1673–1680." U& @6 E6 T* f
0 F% D+ [2 B0 k8 d) C
Greenlee AR, Krisher RL, Plotka ED. Rapid sexing of murine preim-plantation embryos using a nested, multiplex polymerase chain reaction (PCR). Mol Reprod Dev 1998;49:261–267.5 V: J; E" M! C$ p5 A9 j9 ^8 @
7 D. v' G3 I2 `. w0 W& T
Bean CJ, Hassold TJ, Judis L et al. Fertilization in vitro increases non-disjunction during early cleavage divisions in a mouse model system. Hum Reprod 2002;17:2362–2367.6 I- |0 X5 j/ N& }8 `* _  U
* a4 s3 b, k9 |% d, u8 e
Bean CJ, Hunt PA, Millie EA et al. Analysis of a malsegregating mouse Y chromosome: evidence that the earliest cleavage divisions of the mammalian embryo are non-disjunction-prone. Hum Mol Genet 2001;10:963–972.' x7 W, u' T' w/ T# N  G) w
9 k( p3 }4 J% _. g- j; {* E
Simerly C, Dominko T, Navara C et al. Molecular correlates of primate nuclear transfer failures. Science 2003;300:297.
, s- Z/ i& f% `- [7 K# I6 C5 K' ?) {) ^5 v# }9 I9 M
Zuccotti M, Boiani M, Garagna S et al. Analysis of aneuploidy rate in antral and ovulated mouse oocytes during female aging. Mol Reprod Dev 1998;50:305–312.' Y# u$ _# }# ~- I5 L1 h' j

& i: i" F" B1 e6 F" m9 W# N* k  {Boiani M, Eckardt S, Leu AN et al. Pluripotency deficit in mouse clones overcome by clone-clone aggregation: epigenetic complementation? EMBO J 2003;22:5304–5312.
% T( x: V8 ~  L5 L1 D. w% G
0 o5 H% N% B8 ?2 Z. {Handyside AH, Hunter S. A rapid procedure for visualising the inner cell mass and trophectoderm nuclei of mouse blastocysts in situ using poly-nucleotide-specific fluorochromes. J Exp Zool 1984;231:429–434.
( k" k  y0 M1 a4 H# E, ?: r8 `' A- ?! X1 ]7 z* t. |
Boiani M, Eckardt S, Sch?ler HR et al. Oct4 distribution and level in mouse clones: consequences for pluripotency. Genes Dev 2002;16:1209–1219.
3 x* v) w0 p! r& d
* R3 _  Q% F2 N2 @( X- v# R$ tKishikawa H, Wakayama T, Yanagimachi R. Comparison of oocyte-activating agents for mouse cloning. Cloning 1999;1:153–159.% B0 C6 B1 H2 t) J$ ]/ C+ W

! P; T  d+ n) R& P+ ^( QWakayama T, Yanagimachi R. Effect of cytokinesis inhibitors, DMSO and the timing of oocyte activation on mouse cloning using cumulus cell nuclei. Reproduction 2001;122:49–60.
* K! @# Y  c& s$ V+ j! m& P
2 i3 H$ p. `& T5 o% F% DSapienza C, Paquette J, Tran TH et al. Epigenetic and genetic factors affect transgene methylation imprinting. Development 1989;107:165–168.
0 x7 N% V6 {4 u9 }
  t; @3 y1 F- n7 e  W: F4 Q4 gGao S, Czirr E, Chung YG et al. Genetic variation in oocyte phenotype revealed through parthenogenesis and cloning: correlation with differences in pronuclear epigenetic modification. Biol Reprod 2004;70:1162–1170.3 `- Q* J7 N; A1 G% X# @  C

8 K: n. A" z" i6 g* w9 V  V3 GLeppens-Luisier G, Sakkas D. Development, glycolytic activity, and viability of preimplantation mouse embryos subjected to different periods of glucose starvation. Biol Reprod 1997;56:589–596.
& G* S7 n+ N7 U, V
/ G- I( ]1 y2 ]" YScott L, Whittingham DG. Role of facilitative glucose uptake in the glucose-inorganic phosphate-mediated retardation and inhibition of development in different strains of mouse embryos. Reproduction 2002;123:691–700.
, }+ ^( g' N1 u& Z& D
2 Y6 c% ^2 h+ F- o; D7 jNichols J, Zevnik B, Anastassiadis K et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 1998;95:379–391.
7 Q; u, V! C4 {' x5 p3 ^2 o6 c1 k9 \" z0 V) L- f# b
Williams JW, Hawes SM, Patel B et al. Trophectoderm-specific expression of the X-linked Bex1/Rex3 gene in preimplantation stage mouse embryos. Mol Reprod Dev 2002;61:281–287.) A: n9 ]0 K# Y
5 q, f8 f0 ^2 j; }+ T, `- l
Mann MR, Chung YG, Nolen LD et al. Disruption of imprinted gene methylation and expression in cloned preimplantation stage mouse embryos. Biol Reprod 2003;69:902–914.5 L. F: P4 n0 P/ y. G4 s
% F- {5 w3 K1 [; _; j
Gentile L, Monti M, Sebastiano V et al. Single-cell quantitative RT-PCR analysis of Cpt1b and Cpt2 gene expression in mouse antral oocytes and in preimplantation embryos. Cytogenet Genome Res 2004;105:215–221.
% k! ?" L; [8 T' U3 A8 N/ T& J0 n4 i, m5 X) |  M( W# E1 H8 I- z
Latham KE, Patel B, Bautista FD et al. Effects of X chromosome number and parental origin on X-linked gene expression in preimplantation mouse embryos. Biol Reprod 2000;63:64–73.
8 u/ i; U: P* y
  ^& v% V) N, y7 hZeng F, Baldwin DA, Schultz RM. Transcript profiling during preimplantation mouse development. Dev Biol 2004;272:483–496." @0 ^) P) I# `% m8 t1 [" F) ?2 ]
7 P+ s2 e2 C  }
Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 2000;24:372–376.
  e1 b4 [. @3 p
$ _3 d: d2 m* n& SHansis C, Grifo JA, Krey LC. Oct-4 expression in inner cell mass and trophectoderm of human blastocysts. Mol Hum Reprod 2000;6:999–1004.
5 T; B! e, l1 ?! \% Q  t! L; s9 s" ?$ \4 n% n# @2 a% g! d- p/ I1 i0 [/ O; L
Latham KE, Garrels JI, Chang C et al. Quantitative analysis of protein synthesis in mouse embryos. I. Extensive reprogramming at the one- and two-cell stages. Development 1991;112:921–932.7 ]1 I; G0 y! S2 H

8 K" Z* X; p/ [4 kMa J, Svoboda P, Schultz RM et al. Regulation of zygotic gene activation in the preimplantation mouse embryo: global activation and repression of gene expression. Biol Reprod 2001;64:1713–1721.
$ d1 x; t" H8 B! e. D3 ~* i  s$ I
: [( U5 d0 S/ |6 o% fKo MS, Kitchen JR, Wang X et al. Large-scale cDNA analysis reveals phased gene expression patterns during preimplantation mouse development. Development 2000;127:1737–1749.
% S5 s; r5 M4 V- E/ F% E( d
8 G# C9 G& q& YSharov AA, Piao Y, Matoba R et al. Transcriptome analysis of mouse stem cells and early embryos. PLoS Biol 2003;1:E74.
8 |& P7 c6 L7 p' h, y# S% h6 w' Y! ]! }2 B8 }" o0 c
Ho Y, Wigglesworth K, Eppig JJ et al. Preimplantation development of mouse embryos in KSOM: augmentation by amino acids and analysis of gene expression. Mol Reprod Dev 1995;41:232–238.7 b; e9 U3 v2 \6 z; W
  w4 c) s* w8 Z% @
Hamatani T, Carter MG, Sharov AA et al. Dynamics of global gene expression changes during mouse preimplantation development. Dev Cell 2004;6:117–131.9 p6 |5 i% p5 z$ y: w5 N- I/ r
, x' I. y  t( X) L1 q+ V0 `' ], K7 v
Rinaudo P, Schultz RM. Effects of embryo culture on global pattern of gene expression in preimplantation mouse embryos. Reproduction 2004;128:301–311.
- P# c  ^( M3 j  E* J* r9 Z9 M% g& N- C/ P+ }1 a% y
Pain D, Chirn GW, Strassel C et al. Multiple retropseudogenes from pluripotent cell-specific gene expression indicates a potential signature for novel gene identification. J Biol Chem 2005;280:6265–6268.5 f/ y/ |0 c9 {3 f4 b* v! d; k9 O" R

5 H/ ]! k% U7 mYoshimizu T, Sugiyama N, De Felice M et al. Germline-specific expression of the Oct-4/green fluorescent protein (GFP) transgene in mice. Dev Growth Differ 1999;41:675–684.2 ~6 {) Y( X) t8 E5 o' C+ c
8 t. D4 _1 D, N: T. w8 h
Wakayama T, Yanagimachi R. Mouse cloning with nucleus donor cells of different age and type. Mol Reprod Dev 2001;58:376–383.$ ~# k- F9 ]3 X( [: C  _
& b& q  d) r; }+ ^) w. F0 d2 |! m
Doherty AS, Mann MR, Tremblay KD et al. Differential effects of culture on imprinted H19 expression in the preimplantation mouse embryo. Biol Reprod 2000;62:1526–1535.
/ w2 N# ]! O: S2 |5 ?
2 D4 P* \8 [5 y( z" w, @+ Q7 uChung YG, Mann MR, Bartolomei MS et al. Nuclear-cytoplasmic "tug of war" during cloning: effects of somatic cell nuclei on culture medium preferences of preimplantation cloned mouse embryos. Biol Reprod 2002;66:1178–1184.
1 c8 Z" q% k. T
5 w) Q: Z3 o' K& X6 S' _Wakayama T, Perry AC, Zuccotti M et al. Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature 1998;394:369–374.
( F/ O+ x+ N+ _2 U' J( c- A' S0 E# S9 Q3 Y, d
Tada M, Takahama Y, Abe K et al. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr Biol 2001;11:1553–1558.5 e4 i9 p3 e" V

! `3 W* b: A: G- t9 [3 }Do JT, Sch?ler HR. Nuclei of embryonic stem cells reprogram somatic cells. STEM CELLS 2004;22:941–949.
6 G7 M% M$ O' G9 g
' H) Q. s! O6 g) i2 pLusser A, Kadonaga JT. Chromatin remodeling by ATP-dependent molecular machines. Bioessays 2003;25:1192–1200.
* z0 U9 u) [8 H6 a
# k+ s  Y5 G/ p, X6 Z0 DZwaka TP, Thomson JA. A germ cell origin of embryonic stem cells? Development 2005;132:227–233.% O9 }& `1 N$ c2 F: K

& G% z. C$ N, ~, hBlelloch RH, Hochedlinger K, Yamada Y et al. Inaugural article: nuclear cloning of embryonal carcinoma cells. Proc Natl Acad Sci U S A 2004;101:13985–13990.' C* K) `% i0 _: }+ m0 _
$ P8 q, G6 g( r) ]
Humpherys D, Eggan K, Akutsu H et al. Epigenetic instability in ES cells and cloned mice. Science 2001;293:95–97.
& \0 S8 |' Z. J* |7 W1 g7 J# |
* B; I4 `0 i. p% n+ ~! \Hochedlinger K, Jaenisch R. Monoclonal mice generated by nuclear transfer from mature B and T donor cells. Nature 2002;415:1035–1038.- a8 [! C* a5 t2 X

+ ^; B( R8 m2 NLi J, Ishii T, Feinstein P et al. Odorant receptor gene choice is reset by nuclear transfer from mouse olfactory sensory neurons. Nature 2004;428:393–399.
% ?* ^( `( K" ~4 v1 M- ^; H8 k
, k! W: j, m* Q! l1 {Kehler J, Tolkunova E, Koschorz B et al. Oct4 is required for primordial germ cell survival. EMBO Rep 2004;5:1078–1083.
( _! f  k6 i0 R% I0 L+ c5 t
3 G- w4 h9 I+ ?Pickard B, Dean W, Engemann S et al. Epigenetic targeting in the mouse zygote marks DNA for later methylation: a mechanism for maternal effects in development. Mech Dev 2001;103:35–47.- i. J7 H6 ?& d
5 L& K+ |0 t8 G% s' U' K" @6 i0 l$ V
Morin-Kensicki EM, Faust C, LaMantia C et al. Cell and tissue requirements for the gene eed during mouse gastrulation and organogenesis. Genesis 2001;31:142–146.
  o' c0 `3 C2 ]6 w9 h- O7 p: F% y# i* f  N$ k# a6 g
Wilson V, Rashbass P, Beddington RS. Chimeric analysis of T (Brachyury) gene function. Development 1993;117:1321–1331." I& G! {; v1 ^' C2 H' U% f! }

4 s" x& U5 A% V; `3 ZAllen ND, Barton SC, Hilton K et al. A functional analysis of imprinting in parthenogenetic embryonic stem cells. Development 1994;120:1473–1482.
' S$ P! s3 S' r* Y( S2 t7 ]( n) E! o
Mann JR, Gadi I, Harbison ML et al. Androgenetic mouse embryonic stem cells are pluripotent and cause skeletal defects in chimeras: implications for genetic imprinting. Cell 1990;62:251–260.0 P  y  B4 z/ L: N

8 m6 P+ Z! W0 L# F; T0 S1 @9 VParisi T, Beck AR, Rougier N et al. Cyclins E1 and E2 are required for endoreplication in placental trophoblast giant cells. EMBO J 2003;22:4794–4803.. F+ a' W) m# I2 D

  g& o7 m* a. n: ^1 {+ ^7 DWu L, de Bruin A, Saavedra HI et al. Extra-embryonic function of Rb is essential for embryonic development and viability. Nature 2003;421:942–947.
& M" ]9 E$ X0 S; ^8 s& S8 Z
- \( B" s+ z6 }" |7 q* P; l0 N7 L" XMann MR, Lee SS, Doherty AS et al. Selective loss of imprinting in the placenta following preimplantation development in culture. Development 2004;131:3727–3735.
7 i$ j3 Q& b6 v% ~0 g# Y" `& x  C8 P5 b
Ogura A, Inoue K, Ogonuki N et al. Phenotypic effects of somatic cell cloning in the mouse. Cloning Stem Cells 2002;4:397–405.- W7 P' l6 V9 l1 [% A. R3 D
0 R3 z" G6 f2 W6 d! K9 m- \
Brook FA, Gardner RL. The origin and efficient derivation of embryonic stem cells in the mouse. Proc Natl Acad Sci U S A 1997;94:5709–5712.. z9 R- @4 V; [5 a" c
3 |% X7 f! S8 \- p
Chung YG, Ratnam S, Chaillet JR et al. Abnormal regulation of DNA methyltransferase expression in cloned mouse embryos. Biol Reprod 2003;69:146–153.
3 w; @0 o  z4 p) @7 T$ r6 }
3 C1 o$ v: x7 l# L/ [Hattori N, Nishino K, Ko YG et al. Epigenetic control of mouse Oct-4 gene expression in embryonic stem cells and trophoblast stem cells. J Biol Chem 2004;279:17063–17069.
2 t. g$ {: \/ D0 h. \1 G7 |: V6 N, L9 X: a; }: m
Ding S, Schultz PG. A role for chemistry in stem cell biology. Nat Biotechnol 2004;22:833–840.4 e  L) @9 D9 u6 }
/ F1 k8 U, N# I9 b1 C) w
Rosania GR, Chang YT, Perez O etal. Myoseverin, a microtubule-binding molecule with novel cellular effects. Nat Biotechnol 2000;18:304–308.' m$ {. ?% q1 |5 M! ~+ u% ?  c' ^" e
2 u- k+ r3 ^# W7 E3 e( m+ j  m) [
Chen S, Zhang Q, Wu X et al. Dedifferentiation of lineage-committed cells by a small molecule. J Am Chem Soc 2004;126:410–411.7 g- G0 r& B( j2 s

" o3 k1 f, h0 F1 l1 H9 }Shen X, Xiao H, Ranallo R et al. Modulation of ATP-dependent chromatin-remodeling complexes by inositol polyphosphates. Science 2003;299:112–114.
7 u- _$ s/ S  U$ x! _" {( P
/ j3 i$ ~3 D" W. e4 aSteger DJ, Haswell ES, Miller AL et al. Regulation of chromatin remodeling by inositol polyphosphates. Science 2003;299:114–116.
: k5 M' b. w# y* \& [% n0 ^! p6 ~) q( N9 b6 w! d& B5 {1 W8 F
Higgins BD, Kane MT. Inositol transport in mouse oocytes and preimplantation embryos: effects of mouse strain, embryo stage, sodium and the hexose transport inhibitor, phloridzin. Reproduction 2003;125:111–118.2 D' o+ r0 K5 A5 R
3 Z$ k" w! j, p, ~5 }8 y
Burnside AS, Collas P. Induction of Oct-3/4 expression in somatic cells by gap junction-mediated cAMP signaling from blastomeres. Eur J Cell Biol 2002;81:585–591.7 X, t: T. M8 ?# J
) p- L( P5 h+ N( V. o
Eckardt S, Leu NA, Kurosaka S et al. Differential reprogramming of somatic cell nuclei after transfer into mouse cleavage stage blastomeres. Reproduction 2005;129:547–556.(Michele Boiania, Luca Gen)

Rank: 2

积分
56 
威望
56  
包包
1853  
沙发
发表于 2015-6-22 17:26 |只看该作者
知道了 不错~~~  

Rank: 2

积分
64 
威望
64  
包包
1769  
藤椅
发表于 2015-6-25 12:54 |只看该作者
真是天底下好事多多  

Rank: 2

积分
162 
威望
162  
包包
1746  
板凳
发表于 2015-8-3 15:43 |只看该作者
干细胞之家微信公众号
孜孜不倦, 吾等楷模 …………  

Rank: 2

积分
80 
威望
80  
包包
1719  
报纸
发表于 2015-8-11 22:44 |只看该作者
不错,看看。  

Rank: 2

积分
64 
威望
64  
包包
1734  
地板
发表于 2015-8-15 19:38 |只看该作者
楼主福如东海,万寿无疆!  

Rank: 2

积分
68 
威望
68  
包包
1752  
7
发表于 2015-8-21 05:27 |只看该作者
先顶后看  

Rank: 2

积分
162 
威望
162  
包包
1746  
8
发表于 2015-8-22 10:59 |只看该作者
越办越好~~~~~~~~~`  

Rank: 2

积分
64 
威望
64  
包包
1782  
9
发表于 2015-8-24 16:42 |只看该作者
只有一条路不能选择——那就是放弃的路;只有一条路不能拒绝——那就是成长的路。  

Rank: 2

积分
163 
威望
163  
包包
1852  
10
发表于 2015-8-24 18:00 |只看该作者
哎 怎么说那~~  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-4-26 10:31

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.