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: 351468|回复: 241
go

Fibroblast Growth Factor 2 Modulates Transforming Growth Factor Signaling in Mo [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:58 |只看该作者 |倒序浏览 |打印
作者:Boris Greber, Hans Lehrach, James Adjaye作者单位:Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany + n5 ^$ h0 E5 g# J  e" y9 _
                  ; M) B: N4 G( Z: h* R- K1 g
                  7 ^/ M- t1 z; K+ X
          ; k1 @' L) U9 V) s8 H  H
                         8 d* O8 j* Z* |% G0 F2 w  N1 K
            
+ d' x  o5 h6 p: `+ g            
2 V8 n$ D. S5 U            
/ J+ U7 u0 l# l% C' u' K1 s' _7 z3 x* y            ! R2 e3 w' n: Y8 c* p) h" |- R$ m
                      ) G& E) X! |9 t5 Q# t
        
* M& h" ~9 g! i3 z4 I, Q        
: S) a7 H8 o! k+ z2 N% O, \: T1 Y        
& p3 ?, g2 [  e1 Z8 }% }8 s          【摘要】
( }# f, G- G, k8 k$ ]( o% s      Fibroblast growth factor 2 (FGF2) is known to promote self-renewal of human embryonic stem cells (hESCs). In addition, it has been shown that transforming growth factor ß (TGFß) signaling is crucial in that the TGFß/Activin/Nodal branch of the pathway needs to be activated and the bone morphogenic protein (BMP)/GDF branch repressed to prevent differentiation. This holds particularly true for Serum Replacement-based medium containing BMP-like activity. We have reinvestigated a widely used protocol for conditioning hESC medium with mouse embryonic fibroblasts (MEFs). We show that FGF2 acts on MEFs to release supportive factors and reduce differentiation-inducing activity. FGF2 stimulation experiments with supportive and nonsupportive MEFs followed by genome-wide expression profiling revealed that FGF2 regulates the expression of key members of the TGFß pathway, with Inhba, Tgfb1, Grem1, and Bmp4 being the most likely candidates orchestrating the above activities. In addition, restimulation experiments in hESCs combined with global expression analysis revealed downstream targets of FGF2 signaling in these cells. Among these were the same factors previously identified in MEFs, thus suggesting that FGF2, at least in part, promotes self-renewal of hESCs by modulating the expression of TGFß ligands, which, in turn, act on hESCs in a concerted and autocrine manner. " ^6 @7 ~6 I# b- F) [+ A9 ]/ M% X
          【关键词】 Human ESCs Self-renewal Mouse embryonic fibroblasts Basic fibroblast growth factor
) O' D- p$ u. N1 O: m( i: [                  INTRODUCTION! A( u5 Z. m; b  S0 e
8 ]( O) D1 D6 d" A
Human embryonic stem cells (hESCs), like mouse ESCs, are pluripotent and self-renewing cells derived from the inner cell mass of blastocysts. hESCs were originally derived in serum-containing medium and on feeder layers of inactivated mouse embryonic fibroblasts (MEFs) that supported hESCs, allowing them to maintain their undifferentiated state .
# X* f) I& O" I& ]. n  Z; F: _' w) D- h
In addition to LIF-mediated activation of STAT3, mouse ESCs require BMP4 signaling to prevent differentiation to the neural lineage .
. H! [; c- y9 i3 f
( S0 R8 z4 r+ Y5 M/ O# VUsing an inhibitor of ALK 4/5/7 to inactivate SMAD 2/3 signaling, Vallier et al.  presented data suggesting that Activin A induces expression of FGF2 in hESCs. Hence, there is some controversy as to whether FGF2 signaling or the TGFß/Activin/Nodal branch of the TGFß pathway plays the dominant role in sustaining pluripotency of hESCs.
3 K8 s9 y9 W! q/ q) N
% c, _: B$ `: d- i0 @: V" \The widely used protocol for feeder-free hESC culture in MEF-conditioned medium  involves the supplementation with FGF2 both in the conditioning and the hESC culturing steps. We have reinvestigated the rationale for stimulating both MEFs and hESCs with FGF2, which led us to elucidate the immediate downstream targets of FGF2 signaling in both cell types.0 T3 g4 B9 i; ^: n: |

6 i, B& L% g! h/ D' |MATERIALS AND METHODS
7 p0 p* F' Z3 ~! k" g8 S9 M4 d4 W2 E6 y. Y: m  @/ z+ f$ g7 f8 n% K4 z
MEF Cell Culture" E7 u. F. W, L( J2 m$ }. }
2 H/ t% L( g% T7 x2 m+ N
MEFs were isolated according to standard procedures  and 4F-CM, respectively; 0, 4, 13, and 40 ng/ml for FGF2 stimulation experiments). CM was collected daily for a total of 5 days per batch. Before feeding hESCs, 0F-CM and 4F-CM were supplemented with 8 and 4 ng/ml FGF2, respectively, to yield 0F-CM-8F and 4F-CM-4F (Fig. 1B). RNA from FGF2-treated and untreated MEFs was isolated using Qiagen RNeasy kits (Hilden, Germany, http://www1.qiagen.com) with on-column DNase I digestion 1 day after FGF2 stimulation.
+ Z2 H& A2 c0 O( o0 y7 [1 x" w1 s% H$ @9 s2 }
Figure 1. Medium conditioned in the absence of FGF2 fails to sustain human ESC (hESC) self-renewal. (A): Representative morphology of H9 hESCs grown in UM-8F, 4F-CM-4F, and 0F-CM-8F. Scale bar = 0.2 mm. (B): Schematic for the production of 0F-CM-8F versus 4F-CM-4F and morphology of MEFs without and 1 day after FGF2 treatment (4 ng/ml). The same number of cells was plated in both samples. Scale bar = 0.2 mm. (C): Western blot of hESCs grown in the indicated media and probed with OCT4 and GAPDH antibodies. (D): Real-time RT-PCR analysis of hESCs grown in the indicated media using primers for pluripotency markers. Bars indicate SE between biological replicates (0F-CM-8F and UM-8F; n = 2¨C3) or technical variation (4F-CM-4F). Abbreviations: 0F-CM-8F, conditioned medium supplemented with 8 ng/ml FGF2 only after conditioning; 4F-CM-4F, conditioned medium with FGF2 addition before and after the conditioning step; CM, conditioned medium; d, days; FGF2, fibroblast growth factor 2; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; hES, human embryonic stem; MEF, mouse embryonic fibroblast; RT-PCR, reverse transcription-polymerase chain reaction; UM, unconditioned medium; UM-8F, unconditioned medium supplemented with 8 ng/ml FGF2.' Z1 P1 m5 J  l# G" _& N9 d
* n8 Z/ }. M: d- N; G/ u
hESC Culture
) f4 d! E1 q5 J: t& A/ U# O4 u/ Q* f1 d& I: g
H1 and H9 hESCs  were purchased from WiCell Research Institute (Madison, WI, http://www.wicell.org) at passages 45 and 41, respectively, expanded on feeders, adapted to feeder-free culture in MEF-CM for 2 passages, and then used for subsequent experiments. hESCs were confirmed to stain positive for alkaline phosphatase, OCT4, SSEA-4, TRA-1-60, and TRA-1-81 and negative for SSEA-1 (ES Cell Characterization Kit; Chemicon, Temecula, CA, http://www.chemicon.com). hESCs were replated onto six wells precoated with 1 ml of 1/30 (vol/vol) growth factor-reduced Matrigel (BD Biosciences, San Diego, http://www.bdbiosciences.com), using a combination of mechanical and enzymatic passaging. Briefly, 5-day-old colonies were cut into aggregates of 100¨C200 cells using a sterile metal needle before the addition of dispase for 5 minutes (1.5 mg/ml). After extensive washing with UM, the complete contents of the wells were dislodged using a sharp plastic scraper, centrifuged to collect clumps of both undifferentiated and differentiated cells, and replated in the appropriate medium at various splitting ratios (1:2¨C1:4). This was to compensate for reduced plating efficiencies with UM-8F and 0F-CM-8F as compared with 4F-CM-4F. Representative morphology was recorded at a total magnification of x50 using an Olympus CK2 phase contrast microscope (Tokyo, http://www.olympus-global.com). Protein was extracted in Nonidet P40 and Benzonase-containing buffer. RNA was isolated as indicated above.
6 v5 l2 B  o* ^8 B  f2 m! \+ o! @2 ?8 f& C* |( S. ?
Fibroblast-like differentiated cells were derived from H9 hESCs grown in 0F-CM-8F for 3 passages by mechanical removal of remaining undifferentiated colonies. The differentiated cells were further selected by trypsinization and could be expanded in either serum-containing MEF medium (described above), 4F-CM-4F, or¡ªat somewhat lower growth rates¡ªin 0F-CM-8F, but not in unconditioned medium supplemented with or without FGF2 or recombinant insulin-like growth factor 1 (IGF-I) (R&D Systems Inc., Minneapolis, http://www.rndsystems.com). Conditioned medium from human embryonic fibroblast-like (HEF) cells was prepared as with MEFs. For transcriptome comparisons on gene expression chips (described below), we used H9 hESCs grown in conventional MEF-CM, as well as NCCIT embryonal carcinoma cells  (a kind gift of Dr. L. Looijenga) and human foreskin fibroblasts (CRL-2429; American Type Culture Collection, Manassas, VA, http://www.atcc.org) grown in MEF medium.
# z5 R! q$ O" r2 U/ Q
0 D$ E' T2 \( C% ^. |1 \For assaying SMAD2 phosphorylation, H9 hESCs were maintained undifferentiated in 4F-CM-4F for 3 days before exchange to the following media: (a) 4F-CM; (b) 0F-CM; (c) unconditioned medium supplemented with 20 ng/ml recombinant Activin A, 2 ng/ml TGFß1 (in line with Fig. 4E), and 250 ng/ml gremlin 1 (all from R&D Systems); (d) 4F-CM plus 10 µM ALK 4/5/7 inhibitor SB431542 (Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com). All samples were treated in the absence of recombinant FGF2 and with 30 µM SU5402 (Calbiochem, San Diego, http://www.emdbiosciences.com), a potent inhibitor of FGF receptor function , to suppress any FGF signaling in this experiment. Protein was extracted after 12 hours of exposure to the four different media.. t3 b% V' s/ Q: c1 \
1 F+ Q" [/ {* d
In FGF2 restimulation experiments, hESCs were grown in 4F-CM-4F for 2 days, starved in UM without FGF2 and with 30 µM of SU5402 for 24 hours, and, after extensive washing, retreated with 40 ng/ml FGF2 in UM for 4 hours. Controls were FGF2-starved but not restimulated. After the FGF2 starvation period, the cells did not show any obvious signs of spontaneous differentiation, as judged by morphological criteria and monitored by OCT4 immunocytochemistry (supplemental online Fig. 1).. U1 S' v6 x" T9 [

* g. r) w( G0 y9 t- s7 g4 CReal-Time Reverse Transcriptase-Polymerase Chain Reaction, Western Blotting, Immunocytochemistry, and Enzyme-Linked Immunosorbent Assay4 B9 P0 D& M# j1 D7 o  U, U
0 b) g) b7 R, R5 A: `$ d- ?, k
RNA was reverse-transcribed using Moloney murine leukemia virus (USB) and oligo(dT) priming following the manufacturer's instructions. Real-time reverse transcriptase polymerase chain reaction (RT-PCR) was carried out on Applied Biosystems 7900 (Foster City, CA, http://www.appliedbiosystems.com) instrumentation in 20-µl reactions containing 10 µl of SYBR Green PCR mix (ABI), 0.375 µM each primer, and diluted cDNA. All primer pairs used were confirmed to approximately double the amount of product within one cycle and to yield a single product of the predicted size. Primer sequences are provided in supplemental online Table 5. Relative mRNA levels were calculated using the comparative CT method (ABI instruction manual) and presented either as percentage of housekeeping gene expression or as percentage of biological controls. In all cases, results were essentially independent of the gene used for normalization (Gapdh vs. Actb). Where appropriate, statistical significance was evaluated using Student's t test (Figs. 1, 2, 4, *, p & n4 K: u7 j1 x/ y

" T, W& ]0 k! d. mFigure 2. Fibroblast-like differentiated H9 cells support growth of parental human ESCs (hESCs). (A): Morphology of H9-derived HEF cells during expansion. Scale bar = 0.2 mm. (B): Transcriptome comparison between H9 hESCs, hEC cells (line NCCIT), human skin fibroblasts, and HEF cells, based on microarray data; r = linear correlation coefficient. (C): Representative morphology of H9 hESCs grown in medium conditioned by HEF cells. Scale bar = 0.2 mm. (D): OCT4, NANOG, and SOX2 expression level comparison of H9 cells grown in conventional MEF-CM and medium conditioned by HEF cells. Bars indicate standard errors between independent experiments (HEF-CM; n = 3) or technical variation (MEF-CM). Abbreviations: CM, conditioned medium; hEC, human embryonal carcinoma; HEF, human embryonic fibroblast-like; hES, human embryonic stem; MEF, mouse embryonic fibroblast; RT-PCR, reverse transcription-polymerase chain reaction.
. T3 [/ i: i1 C6 `' ]( {& v. X+ |% M5 ~
Western blotting was performed on 10¨C30-µg protein samples separated by SDS-polyacrylamide gel electrophoresis and transferred onto nitrocellulose membranes. Primary antibodies used were Santa Cruz Biotechnology sc-8629 (1/5,000; Santa Cruz, CA, http://www.scbt.com), Ambion 4300 (1/5,000; Austin, TX, http://www.ambion.com), and Cell Signaling Technology 3102 and 3108 (both at 1/1,000; Beverly, MA, http://www.cellsignal.com). Appropriate peroxidase-conjugated secondary antibodies in conjunction with enhanced chemiluminescence detection kits and films (Amersham Biosciences, Piscataway, NJ, http://www.amersham.com) were used for visualization. OCT4 immunostaining was performed using antibodies sc-8629 (1/100; Santa Cruz Biotechnology) and A21468  (1/300; Amersham Biosciences). Enzyme-linked immunosorbent assays (ELISAs) were carried out using products 338-AC, MAB3381, BAM3381, MB100B, and DY998 from R&D Systems, following the manufacturer's recommendations. The linear working range of the Activin A assay was between 0.25 and 32 ng/ml.! h9 x% b: g% a0 S3 s6 e" z% o

. {5 i$ t( H. ^: P; w9 qChip Hybridizations and Analysis of Whole-Genome Expression Data; Y, X* s! V. N& m

3 X4 T; ^+ c  `# R) X+ z' v6 p7 NBiotin-labeled cRNA was produced by means of a linear amplification kit (IL1791; Ambion, Austin, TX, http://www.ambion.com) using 300 ng of quality-checked total RNA as input. Chip hybridizations, washing, Cy3-streptavidin (Amersham Biosciences) staining, and scanning were performed on an Illumina BeadStation 500 (San Diego, http://www.illumina.com) platform using reagents and following protocols supplied by the manufacturer. cRNA samples were hybridized on Illumina mouse-6 (CF1 MEFs), mouse-8 (NMRI and CD1 MEFs), and human-8 (for all human samples) BeadChips. The mouse-8 and human-8 chips cover approximately 24,000 RefSeq transcripts, whereas the mouse-6 format contains an additional 23,000 features. Annotation information for the individual chip formats is publicly available from Illumina. The inherent manufacturing principle to randomly distribute large populations of oligonucleotide-coated beads across the available positions enables 30 intensity measurements per feature on average, yielding quantitative results  (p 3 a  o5 g" _. e/ u3 U

+ q8 t6 h( N% Y9 H! R* c5 cRESULTS% ?3 g- z7 ]( C: M; |
& X1 z( {# C. T- I# E/ Q
Extrinsic FGF2 Signaling Is Crucial for the Production of Effective MEF-Conditioned Medium
1 i& X3 k* O( p, |2 d4 ?5 _; ^8 p, X2 `6 _! J4 d+ w
According to Xu et al.  including 4 ng/ml FGF2 on high-density cultures of inactivated MEFs (4F-CM). Before feeding hESCs grown on Matrigel-coated plates, another 4 ng/ml FGF2 is added (4F-CM-4F). To investigate the importance of supplementing the medium with FGF2 as early as in the conditioning step, an alternative medium was prepared, termed 0F-CM-8F, in which FGF2 was added to a final concentration of 8 ng/ml only after the conditioning step (Fig. 1B). While preparing the two types of conditioned media, we noticed that FGF2 induced a morphological change in the MEFs, yielding more elongated/stringy cells within 1 day (Fig. 1B). We then compared the ability of 4F-CM-4F and 0F-CM-8F to support the undifferentiated growth of H9 hESCs on Matrigel-coated plates. As a control, we also used unconditioned medium supplemented with 8 ng/ml FGF2 (UM-8F). Within 5 days of culture, morphological differences became apparent. H9 cells grown in 4F-CM-4F were almost exclusively forming round and tightly packed colonies with rather defined boundaries, indicative of undifferentiated growth. In contrast, colonies grown in 0F-CM-8F showed differentiation from the inside of the colonies in more than 50% of cases, as well as a fibroblast-like type of differentiation at the colonies' edges. Cells grown in UM-8F formed rather irregularly shaped and relatively loose colonies with differentiation predominantly at the colonies' boundaries (Fig. 1A). Upon further passaging (5 days per passage), colonies grown in 4F-CM-4F and UM-8F retained their morphology; however, the plating efficiencies in UM-8F were significantly reduced compared with 4F-CM-4F. In contrast, at passage 3, the numbers and average sizes of colonies in 0F-CM-8F were further reduced, and the fibroblast-like differentiated cells occupied almost the entire surface area between the colonies. These, however, now looked tightly packed and had defined boundaries (Fig. 1A). We monitored these morphological differences between cells grown in 4F-CM-4F versus 0F-CM-8F by Western blotting using an antibody against the pluripotency marker OCT4. Indeed, the OCT4 level was strongly reduced in 0F-CM-8F as compared with conventional CM (Fig. 1C). The morphological differences described above were reproducible between independent experiments. We routinely quantified the expression levels of OCT4 and two additional pluripotency markers (NANOG and SOX2) using real-time RT-PCR in cells cultured with 4F-CM-4F, 0F-CM-8F, and UM-8F for 3 passages. In line with our morphological observations, the expression levels of all three marker genes were strongly reduced in 0F-CM-8F, to levels similar to those in UM-8F (Fig. 1D). The following conclusions/hypotheses can be drawn from these data:
7 ^( i& @/ ?: n( j5 x
- N5 z0 |" `( P+ m% u" t/ B" TFGF2 stimulates the secretion of one or more supportive factors in MEFs (compare 4F-CM-4F vs. UM-8F and 0F-CM-8F in Fig. 1A, 1B, 1D).2 S9 s/ X' n' H- _) p

3 q+ H3 l( F% f. z. O0F-CM-8F contains enhanced levels of differentiation-inducing activity (compare 0F-CM-8F vs. UM-8F and 4F-CM-4F in Fig. 1A, 1D).
. d( ^7 C( E3 B/ M7 M
( ~6 g# o5 K, _9 c* j, ?The fibroblast-like cells appearing with 0F-CM-8F upon continuous passaging support the remaining undifferentiated hESCs (compare morphology with 0F-CM-8F, passage 3 vs. passage 1 in Fig. 1A).' H/ _3 \2 |+ R* B, D4 f% ]4 w

% Q+ H7 S' f& U9 V0 yFibroblast-Like Cells Emerging in 0F-CM-8F Support Remaining Undifferentiated hESCs
( l/ h/ n1 M1 X4 k1 L) \* r$ U* [/ y. P3 `; |% N2 K2 f/ r
To test the hypothesis 3, the remaining undifferentiated colonies from passage 3 0F-CM-8F cultures were mechanically removed, and the fibroblast-like human cells expanded without an apparent change in morphology (Fig. 2A). We profiled the transcriptome of these cells on whole-genome expression chips, along with samples of undifferentiated H9 cells, NCCIT human embryonal carcinoma (hEC) cells, and human skin fibroblasts. A correlation-based clustering analysis showed that the H9 derivative cells were very similar to the control human fibroblast line, whereas the undifferentiated parental hESCs clustered together with the hEC cell line (Fig. 2B). Hence, we termed the H9 derivatives HEF cells, in accordance with published work . Furthermore, we inactivated these cells for the production of conditioned hES medium. CM produced with HEF cells was able to support the parental cells throughout the tested time range (4 passages). The colonies appeared tightly packed and overall undifferentiated, with some spontaneous differentiation at the edges (Fig. 2C). Real-time PCR of OCT4, NANOG, and SOX2 expression levels was carried out to show that HEF-based CM was almost equally efficient as MEF-CM in maintaining the undifferentiated state of hESCs (Fig. 2D). Taken together, these data suggest that the fibroblast-like differentiated cells emanating from 0F-CM-8F cultures upon continuous passaging (Fig. 1A) do indeed support the remaining undifferentiated cells, which partially compensated for the otherwise nonsupportive character of the 0F-CM-8F medium.
8 o1 E' l( @, S: s  e3 x" o
0 L1 I: H% D1 U) X% lFGF2 Regulates Key Members of the TGFß Pathway in MEFs" ]6 ~. T/ X  |$ |  J! w6 k& d
( G( h6 @4 ]; q7 m8 O
To address conclusions/hypotheses 1 and 2 above, inactivated MEFs of mouse strain CF1 were treated with various doses (0, 4, 13, and 40 ng/ml) of FGF2 in unconditioned hES medium for 24 hours, and global expression analysis was carried out (supplemental online Table 1). Overall, FGF2 treatment caused differential expression of several thousand genes in MEFs. Those with an at least twofold significant change in expression between the FGF2-treated samples and the 0 ng/ml controls were filtered on the basis of Gene Ontology terms "extracellular space" (GO:0005615) and "receptor-binding" (GO:0005102) . This analysis confirmed that members of the TGFß pathway were significantly overrepresented among the secreted factors regulated by FGF2 (p
8 T1 Q& \1 s- G  Y; m. W+ K/ X6 H
1 q. ^  i4 ^5 N1 h4 zFigure 3. Global gene expression analysis of FGF2-stimulated versus nontreated MEFs. (A): Expression data of treated samples plotted against untreated ones (n = 3). Small dots represent differentially expressed genes (p 1 w# {2 d' `, `3 s+ R$ ~
, r8 |6 S) ^8 F8 ]/ Q
Tgfb1, Inhba, Grem1, and Bmp4 Are the Genes Expressed in MEFs Most Likely To Influence hESC Self-Renewal( J3 f! `; t" a( I+ c
: R6 m' ?. @5 v
To further confirm the above results, we used a different hESC line, H1, as well as additional batches of MEFs derived from mouse strains other than the widely used CF1, namely NMRI and CD1. To rule out an influence of the H1 passage number, we compared in parallel the performances of CF1-, NMRI-, and CD1-conditioned medium prepared with or without FGF2 supplementation. As before, we obtained early trophoblast-like differentiation in the centers of the colonies , as well as differentiation around the colonies after several days in 0F-CM-8F with all three batches of MEFs, whereas H1 cells grown in CF1 and NMRI 4F-CM-4F remained undifferentiated throughout the experiment. Somewhat surprising was the fact that hESCs in CD1 4F-CM-4F differentiated partially, almost to a similar extent as the cells in CF1 and NMRI 4F-CM-4F, but still less than in 0F-CM-8F from the same batch of MEFs (representative morphologies are given in Fig. 4A). To quantify these observations, mRNA expression levels of the core hESC transcription factors OCT4, NANOG, and SOX2 were again recorded using real-time PCR at passage 2 (Fig. 4B). The lack of support by the CD1 4F-CM-4F medium in comparison with the two supportive batches of CF1 and NMRI feeders was confirmed using H9 hESCs in additional experiments (results not shown).( U% Q4 x; X! Z; ~, e+ k& k

: Z  n3 O* U; _0 l& i& [8 \4 PFigure 4. Nonsupportive MEFs may be used to narrow down candidate genes relevant for human ESC (hESC) self-renewal. (A): H1 hESCs differentiated rapidly in conditioned medium with FGF2 addition before and after the conditioning step (4F-CM-4F) produced from nonsupportive feeders and in conditioned medium supplemented with 8 ng/ml FGF2 only after conditioning (0F-CM-8F) of all MEF batches tested. Representative pictures were taken from hESC cultures fed with the indicated media for 5 days. Scale bar = 0.2 mm. (B): Real-time reverse transcription-polymerase chain reaction (RT-PCR) analyses on samples in (A) at the end of passage 2 (10 days) using primers for pluripotency markers. Note the low levels of OCT4, NANOG, and SOX2 expression in CD1 4F-CM-4F compared with the two supportive MEF strains. Bars indicate technical standard errors. (C): Venn diagram illustrating a subtractive approach to filtering microarray data for genes encoding secreted proteins that are differentially expressed in supportive MEFs (overlap of CF1 and NMRI chip data) in response to FGF2 but not in the nonsupportive feeders. The 42 genes, notably encoding both soluble proteins and constituents of the extracellular matrix, are listed in supplemental online Table 3. (D): Real-time RT-PCR analyses of supportive and nonsupportive MEFs treated for 1 day with different doses of FGF2 using primers for upregulated TGFß pathway members from supplemental online Table 3 and Bmp4. Note that Bmp4 was downregulated in all cases, but its expression level (as a percentage of Actb) was substantially higher in nonsupportive MEFs compared with those in CF1 and NMRI fibroblasts. The charts are drawn on the same scale to better illustrate this fact. (E): Enzyme-linked immunosorbent assay (ELISA)-based measurements of Activin A and TGFß1 concentrations in 0F-CM and 4F-CM prepared with CF1 and CD1 fibroblasts. In contrast to (D) (1 day of FGF2 treatment), CM was collected for 5 days and then pooled. NMRI cells from the same batch used in (A¨CD) were no longer available at the time the ELISAs were conducted. Error bars denote standard deviations between technical replicates (n = 2). (F): Western blot showing decreased SMAD2 phosphorylation (at Ser465/467) in 0F-CM versus 4F-CM from supportive feeders. Total SMAD 2/3 served as loading control. Note that loading of this sample was somewhat lower than in lanes 1 and 2. Abbreviations: 0F-CM-8F, conditioned medium supplemented with 8 ng/ml FGF2 only after conditioning; 4F-CM-4F, conditioned medium with FGF2 addition before and after the conditioning step; 4F-CMSB, 4F-CM plus ALK 4/5/7 inhibitor SB431542; Bmp, bone morphogenic protein; FGF2, fibroblast growth factor 2; MEF, mouse embryonic fibroblast; TGF, transforming growth factor; UMGTA, unconditioned medium supplemented with recombinant gremlin 1, TGFß1, and Activin A to mimic 4F-CM.3 `  a6 X6 T8 S1 Y  D0 Y

$ q+ Z5 v; \# S  C9 |0 IWe therefore reasoned that the nonsupportive MEFs could be used to narrow down the list of the previously identified TGFß candidate genes. To this end, RNA samples from FGF2-stimulated and nontreated MEFs of the second supportive strain and the nonsupportive strains were again hybridized on expression arrays, and differentially expressed genes encoding secreted proteins were identified as before. We then devised a subtractive approach, illustrated in Figure 4C, to select genes encoding secreted factors that are differentially expressed upon FGF2 treatment in both supportive batches of MEFs but not in the nonsupportive ones (supplemental online Table 3). Interestingly, this list included two genes encoding SMAD 2/3 activators, Tgfb1 and Inhba, and one gene coding for a BMP4 antagonist, Grem1. The primary candidate for differentiation-inducing activity, Bmp4, appeared to be significantly downregulated upon FGF2 treatment in all three MEF batches. However, we noticed that the Bmp4 expression levels were considerably higher in the nonsupportive MEFs, according to the array data. Therefore, real-time confirmations at several FGF2 concentrations were carried out on Tgfb1, Inhba, Grem1, as well as on Bmp4 (Fig. 4D). Indeed, using this alternative technique for mRNA level quantification, Tgfb1, Inhba, and Grem1 were strongly upregulated in a concentration-dependent manner upon FGF2 treatment, using MEFs of the two supportive strains, whereas the expression levels of these genes did not change or were only slightly induced in the nonsupportive feeder cells. In contrast, Bmp4 expression levels dropped substantially with increasing doses of FGF2 in all three MEF batches. However, the overall Bmp4 mRNA levels in the nonsupportive MEFs were substantially higher, especially at moderate concentrations of FGF2 (4 and 13 ng/ml; Fig. 4D).
9 F- U# `, G  B1 N
! N0 c( a% \" U) T1 u* fTo test whether these changes in mRNA abundance also become apparent at the protein level, we performed ELISA-based analyses of Activin A and TGFß1 concentrations in 0F-CM and 4F-CM produced with supportive and nonsupportive MEFs. As shown in Figure 4E, Activin A and TGFß1 protein levels were approximately 10 times higher in conditioned medium produced from FGF2-stimulated CF1 feeders as compared with 0F-CM of the same cells. In contrast, TGFß1 and in particular Activin A levels were substantially lower in 4F-CM prepared with nonsupportive CD1 MEFs.% z- @3 ^1 f8 f9 E5 e8 R

  [- ~1 A# a9 p3 aThese data suggest that 4F-CM-4F supports hESC self-renewal via activation of SMAD 2/3, in contrast to 0F-CM-8F, which contains only low levels of Activin A and TGFß1. We sought to demonstrate this by assaying the SMAD2 phosphorylation status in response to the two types of conditioned medium, under FGF2 starving conditions in hESCs. As shown in Figure 4F, SMAD2 phosphorylation was substantially enhanced in 4F-CM as compared with 0F-CM, and this effect could be mimicked by extrinsic supplementation of unconditioned medium with recombinant Activin A, TGFß1, and gremlin 1. In cells treated with 4F-CM and an inhibitor of TGFß/Activin/Nodal type I receptors, SMAD2 phosphorylation was abolished, as expected.5 k" p4 U# j  ^, s2 K6 [

2 s* s# S8 @& P& ]# E1 R/ Y( EDownstream Targets of FGF2 Signaling in hESCs
4 g1 G% U6 J' H+ S* G- V5 \! y$ x9 B! S
According to Xu et al. . We further annotated differentially expressed genes with Gene Ontology terms to reveal overrepresented functional categories. Interestingly, there were more than 30 genes involved in cell cycle regulation (GO:0007049; supplemental online Table 4), suggesting that FGF2 is regulating genes associated with hESC proliferation, as well as more than 40 genes encoding transcription factors (GO:0003700; supplemental online Table 4), revealing complex downstream effects of FGF2 signaling in hESCs. Transcription factors upregulated in response to FGF2 stimulation included zinc finger proteins, SRY- and forkhead box TFs (SOX4, -8, and -13; FOXC2, -D1, and -L2), cellular oncogenes (FOS, JUN, MYC, and ETS2), and, interestingly, the pluripotency controlling gene NANOG.( X/ q9 R" N$ L0 g

5 ^# D& d7 q7 y" O+ h. A9 O* @Moreover, the lists of up- and downregulated FGF2 targets were mapped to known signaling pathways. Using high statistical stringency (p 8 M9 @% ]2 U6 t' ~9 L

* p1 K* W+ b6 r* j4 `Figure 5. FGF2 regulates key TGFß members in both MEFs and hESCs, resulting in support of the undifferentiated state. (A): Real-time reverse transcription-polymerase chain reaction confirmations of selected genes differentially expressed in response to exogenous FGF2 in hESCs. Array-based expression ratios from supplemental online Table 4 are also shown for comparison. Note the logarithmic scale of the fold changes and the high agreement between the array- and real-time-based measurements. The array oligo for TGFB1 was found to be falsely designed. Error bars indicate variation between two independent restimulation experiments. (B): Simplified scheme of the TGFß pathway modified from . Differential expression of the Nodal gene was observed neither in MEFs nor in hESCs. (C): Model of regulatory relationships between FGF2, TGFß pathway, and core hESC transcription factors, integrating results from the present study and published data (supporting evidence given in Discussion). Arrows denote induction or repression of gene transcription. Abbreviations: av., average; BMP, bone morphogenic protein; FGF2, fibroblast growth factor 2; hES, human embryonic stem; hESC, human ESC; MEF, mouse embryonic fibroblast; TGFB1, transforming growth factor ß1.5 G+ W0 x1 a- r& P% l" a7 @& d
3 z! l/ P2 E  Q; B* |7 a! Y, f
DISCUSSION
/ }2 r* |( M; c$ g7 o% {, W# O/ S: c# a: \" \/ e4 D- W
Our reinvestigation of a widely used protocol for feeder-free hESC culture  described the derivation of a similar if not identical cell type termed HEF1 via the formation of embryoid bodies and subsequent selection in serum-containing medium. Growing hESCs in 0F-CM-8F for several passages followed by mechanical removal of the few remaining undifferentiated colonies would simplify the derivation and initial scale-up culture of supportive human feeder cells from any parental line, if desired.2 g# t& M+ Y$ Z+ Z

" Y& ~$ ?) j4 XFGF2 stimulation of MEFs leads to a morphological change sometimes falsely attributed to the Serum Replacement contained in basic hESC medium (personal communication) and induced overt differential gene expression (Fig. 3A). Regarding the feeder-free growth of hESCs on Matrigel, only genes encoding soluble secreted proteins appeared to be of relevance in this study. We therefore made use of Gene Ontology annotation to filter the large set of genes differentially expressed in response to FGF2. In a stepwise subtractive approach using supportive and nonsupportive batches of MEFs, the list of candidate genes conferring beneficial or detrimental activities in the context of hESC self-renewal was narrowed down essentially to Tgfb1, Inhba, Grem1 (all upregulated by FGF2), and Bmp4 (downregulated), respectively. The effects of the encoded factors on hESC self-renewal are well documented: TGFß1 and Activin A prevent differentiation via activation of SMAD 2/3 .
+ _- i. H4 m9 Q; y  \* T( a% N! }: J* a' N, H& e# m1 J
There may be additional factors released by MEFs that have an impact on hESC self-renewal. For instance, Igf1 as a candidate for promoting proliferation of differentiated fibroblast-like cells was strongly downregulated in response to FGF2 (supplemental online Table 2), whereas Wnt5a was upregulated exclusively in supportive feeders (supplemental online Table 3). However, preliminary experiments in which we tested recombinant forms of the encoded proteins (0¨C100 ng/ml) for effects on hESC proliferation and/or differentiation did not reveal evident results (not shown). Pyle et al. .( E2 o" |; |  Q6 R0 z  d) W
$ m$ [. n3 Y3 G2 y9 k" ?  g! v, `8 Z
To test whether FGF2 also regulates key members of the TGFß pathway in hESCs and to reveal overall downstream targets of FGF2 signaling, restimulation experiments were performed followed by whole-genome expression analysis. The upregulation of known FGF2 target genes and the high degree of consistency with real-time RT-PCR validations suggests that the data set is of high quality (Fig. 5A). To our knowledge, such data has not yet been published and will therefore present a useful basis for further investigation in the field. The gene set presented in supplemental online Table 4 includes large numbers of transcription factors, genes with unknown functions, and additional genes of potential interest such as GJA7 and GJB2 (both upregulated) encoding gap junction proteins. This is noteworthy because functional gap junctions have been shown to be present in undifferentiated hESCs  (Fig. 5B)./ i) E3 b3 t" k6 ?3 S+ q0 a
. C7 x$ F' K3 u$ {- E8 |
Notably, there is some indication of these secondary effects, in that ID1, a known downstream target of BMP signaling .! L. m2 G; J1 r# e0 ^) p
- @$ j8 E; H" d! B: I( m0 B0 M9 N/ Y
In SR-based medium containing BMP-like activity, either a very high dose of exogenous FGF2  found that even low levels of BMP signaling may be required to prevent differentiation to neurectoderm. In conclusion, FGF2 appears to be the most upstream self-renewal factor in human embryonic stem cells.% a9 S* n* s+ s! Q) r
) }) y+ V8 S& F$ v, H
DISCLOSURES
( M' S3 {1 i3 E% W) C8 }3 d) r! X3 O0 a$ i7 h' q+ G
The authors indicate no potential conflicts of interest.
% l. \5 ]4 C" N& _- G  `) ?* b) f) U, H( X2 A, ?9 o: t
ACKNOWLEDGMENTS9 x. k9 o7 b3 i) f0 k
# i" I  X! b5 Y8 {! p
We are grateful to Drs. Tom Burdon, Wei Cui, and Judy Fletcher for sharing their knowledge of hESC culture. This work was supported by Deutsche Forschungsgemeinschaft Grant DFG-AD 184/4-1.3 k0 q2 U6 W7 U, u( l
          【参考文献】
4 y  M0 @; R" |. I, A  M
2 g4 d: L" {$ J: I; r/ Q7 f
( A! p0 G$ y6 e: y$ {. S) wThomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145¨C1147.% k1 T- g- S# v3 k5 }4 S
1 R2 u7 z: Q/ u! J$ L' n& u
Price PJ, Goldsborough MD, Tilkins ML Invitrogen. Embryonic stem cell serum replacement. International patent WO9830679 7 16, 1998;.+ T+ m7 o! v0 `5 s# T

. Y- b( m% T1 V5 L# ]/ u5 qAmit M, Carpenter MK, Inokuma MS et al. Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture. Dev Biol 2000;227:271¨C278.
  Z# |! A( M2 p4 x) l8 @, w" m" J  C+ y6 t* Q& [
Xu C, Inokuma MS, Denham J et al. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol 2001;19:971¨C974.
2 D' h6 p7 a3 K) V& u: l  a+ h% n! M+ S4 I9 S' V3 y" h& J
Kleinman HK, McGarvey ML, Hassell JR et al. Basement membrane complexes with biological activity. Biochemistry 1986;25:312¨C318.$ |5 d2 ~! {4 o9 |+ s- o

0 ^# q$ a7 }. ]: e8 GLudwig TE, Levenstein ME, Jones JM et al. Derivation of human embryonic stem cells in defined conditions. Nat Biotechnol 2006;24:185¨C187.
+ o* Z, _; ~7 ]+ J0 e
! r+ X1 X% b3 `! e9 Y) s- ~' hLu J, Hou R, Booth CJ et al. Defined culture conditions of human embryonic stem cells. Proc Natl Acad Sci U S A 2006;103:5688¨C5693.8 K5 @, n( D1 L9 u
& p& r* E" v5 h# v- k
Yao S, Chen S, Clark J et al. Long-term self-renewal and directed differentiation of human embryonic stem cells in chemically defined conditions. Proc Natl Acad Sci U S A 2006;103:6907¨C6912.
8 f' A  l( l6 E
0 T& j$ G8 _& [; j0 p3 B/ JChambers I, Smith A. Self-renewal of teratocarcinoma and embryonic stem cells. Oncogene 2004;23:7150¨C7160.
9 {& `& r7 m& s* t$ ]% Z6 g* x- v% ]2 N) b% |9 F* g
Ying QL, Nichols J, Chambers I et al. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell 2003;115:281¨C292.
, H5 M/ A/ b: M* t' {. l7 o- Z* c! v* ~5 P0 ?: H, }
Daheron L, Opitz SL, Zaehres H et al. LIF/STAT3 signaling fails to maintain self-renewal of human embryonic stem cells. STEM CELLS 2004;22:770¨C778.- Z: E& _' m: X4 c' m2 Q& y% m8 R, e) M6 j
. S  _1 g* W- [9 U  j
Humphrey RK, Beattie GM, Lopez AD et al. Maintenance of pluripotency in human embryonic stem cells is STAT3 independent. STEM CELLS 2004;22:522¨C530.
" h: }5 f$ B( H! ^7 I
$ J! M  Q+ @5 OXu RH, Chen X, Li DS et al. BMP4 initiates human embryonic stem cell differentiation to trophoblast. Nat Biotechnol 2002;20:1261¨C1264./ J8 O! d0 ~6 ?+ [, a4 L4 m3 \
+ ^$ P! m0 V% n" d' q6 ~- H( f- n: S
Pera MF, Andrade J, Houssami S et al. Regulation of human embryonic stem cell differentiation by BMP-2 and its antagonist noggin. J Cell Sci 2004;117:1269¨C1280.
2 f7 D% n6 F$ S( A; K' W' C9 x; }2 \# \! y  H; k
Xu RH, Peck RM, Li DS et al. Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells. Nat Methods 2005;2:185¨C190.
( Z) ~+ y% R* d9 x4 ]: W! q9 Z% z/ |% A  m! _
Wang G, Zhang H, Zhao Y et al. Noggin and bFGF cooperate to maintain the pluripotency of human embryonic stem cells in the absence of feeder layers. Biochem Biophys Res Commun 2005;330:934¨C942.
: t* w: u# i$ G" M" s% W; i; I; l
Valdimarsdottir G, Mummery C. Functions of the TGFbeta superfamily in human embryonic stem cells6 T7 p  o+ D: J' k% N- P

9 p9 ^6 P0 M4 w6 L+ Q8 |& ?James D, Levine AJ, Besser D et al. TGFbeta/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development 2005;132:1273¨C1282.
: s* n, j5 ?2 M! c
" ~* r1 t6 f# x7 ZBeattie GM, Lopez AD, Bucay N et al. Activin A maintains pluripotency of human embryonic stem cells in the absence of feeder layers. STEM CELLS 2005;23:489¨C495.2 a4 O# Q' @2 I* r6 r; g

( ]9 R& A! ~+ H2 G3 dXiao L, Yuan X, Sharkis SJ. Activin a maintains self-renewal and regulates fibroblast growth factor, wnt, and bone morphogenic protein pathways in human embryonic stem cells. STEM CELLS 2006;24:1476¨C1486.# q: i0 T0 v+ {2 O+ y1 Z9 o1 k7 o

6 s: Z$ ^- k$ I5 @& y9 |Vallier L, Reynolds D, Pedersen RA. Nodal inhibits differentiation of human embryonic stem cells along the neuroectodermal default pathway. Dev Biol 2004;275:403¨C421.
8 o0 b* ^+ B' J% s; b2 M) f% F2 R* [  F$ q* z" `$ o: k: h
Amit M, Shariki C, Margulets V et al. Feeder layer- and serum-free culture of human embryonic stem cells. Biol Reprod 2004;70:837¨C845.
, E/ q& E4 g, j; v# u; h- @: J
Poon E, Clermont F, Firpo MT et al. TGFbeta inhibition of yolk-sac-like differentiation of human embryonic stem-cell-derived embryoid bodies illustrates differences between early mouse and human development. J Cell Sci 2006;119:759¨C768.
, _: P) j, r! G7 [  }/ E
* r! ^6 M% G5 L0 D) D$ t: IVallier L, Alexander M, Pedersen RA. Activin/Nodal and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells. J Cell Sci 2005;118:4495¨C4509.
0 r0 R" l7 q; g' g: h, X- a0 @. s4 c. Y9 h
Nagy A, Gertsenstein M, Vintersten K et al. Manipulating the Mouse Embryo: A Laboratory Manual. 3rd ed. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press,2003;371¨C373.
, M: o9 U- f+ I2 k  W8 o+ I% f, w0 V6 R3 k' d0 ^/ y
Teshima S, Shimosato Y, Hirohashi S et al. Four new human germ cell tumor cell lines. Lab Invest 1988;59:328¨C336.9 f7 W0 x7 x7 R- K, D7 y

7 b" V6 a& {* P% z. L+ T) IMohammadi M, McMahon G, Sun L et al. Structures of the tyrosine kinase domain of fibroblast growth factor receptor in complex with inhibitors. Science 1997;276:955¨C960.% u# L1 f4 s* N7 s
- r% M0 t1 X% i
Kuhn K, Baker SC, Chudin E et al. A novel, high-performance random array platform for quantitative gene expression profiling. Genome Res 2004;14:2347¨C2356.1 d# a9 x3 Y: o% @' L7 t, w
9 f4 G$ Y2 E6 j0 b: z. H0 o
Xu C, Jiang J, Sottile V et al. Immortalized fibroblast-like cells derived from human embryonic stem cells support undifferentiated cell growth. STEM CELLS 2004;22:972¨C980.
) K! H7 }# P1 B- ]7 Y. J% a  g+ Q+ r
0 s' N+ H( B( u( M: `Ashburner M, Ball CA, Blake JA et al. Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 2000;25:25¨C29.
, L) `& z; V* y2 X6 c( b2 k+ L( @" I1 f5 X; y( g
Kanehisa M. A database for post-genome analysis. Trends Genet 1997;13:375¨C376.
: P( N8 x  {8 @9 \1 d' Z# E& h& K/ c% F6 Y6 r# [
Dennis G Jr, Sherman BT, Hosack DA et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol 2003;4:P3.
8 W! x+ r  M; k* u" W% a' a2 f
; A2 h" b5 o. [, _, o2 ~Dvorak P, Dvorakova D, Koskova S et al. Expression and potential role of fibroblast growth factor 2 and its receptors in human embryonic stem cells. STEM CELLS 2005;23:1200¨C1211.
! e8 n! o* F9 j/ @7 P9 `) q/ `( V% f$ r
Kang HB, Kim JS, Kwon HJ et al. Basic fibroblast growth factor activates ERK and induces c-fos in human embryonic stem cell line MizhES1. STEM CELLS Dev 2005;14:395¨C401.
$ H3 k: V$ A3 i. y$ ?4 V% @+ l1 e/ R2 W
Wakioka T, Sasaki A, Kato R et al. Spred is a Sprouty-related suppressor of Ras signalling. Nature 2001;412:647¨C651.
- ~/ ?+ j$ M" R1 b8 X0 W# E% A- o$ @& _$ u! f0 A( u- w* P" o
Kim HJ, Bar-Sagi D. Modulation of signalling by Sprouty: A developing story. Nat Rev Mol Cell Biol 2004;5:441¨C450.
; E  Z2 v; M+ a4 W7 ~3 Z% \- j1 S0 ^7 [  a6 |  f7 C
Pyle AD, Lock LF, Donovan PJ. Neurotrophins mediate human embryonic stem cell survival. Nat Biotechnol 2006;24:344¨C350.7 B1 @7 n: w2 O( R) {; \

% h+ \; h* B& g2 k: z  T# ZCarpenter MK, Rosler ES, Fisk GJ et al. Properties of four human embryonic stem cell lines maintained in a feeder-free culture system. Dev Dyn 2004;229:243¨C258.
% v$ x; V4 S% B$ M
! S) Z0 t' d$ u; T+ b( SWong RC, Pebay A, Nguyen LT et al. Presence of functional gap junctions in human embryonic stem cells. STEM CELLS 2004;22:883¨C889.
1 U4 K/ N3 W, c' m# H1 k7 }7 U+ Y6 _( f; |. e
Huettner JE, Lu A, Qu Y et al. Gap junctions and connexon hemichannels in human embryonic stem cells. STEM CELLS 2006;24:1654¨C1667.
# A6 D  v. G* K6 i: \  s! B' N1 M: r' @1 b0 ?
Belo JA, Bachiller D, Agius E et al. Cerberus-like is a secreted BMP and nodal antagonist not essential for mouse development. Genesis 2000;26:265¨C270.# Q5 `* @0 \3 {2 ?
! J( x2 m* h# K9 n& r: W
Bhattacharya B, Miura T, Brandenberger R et al. Gene expression in human embryonic stem cell lines: Unique molecular signature. Blood 2004;103:2956¨C2964.
% @! }2 _+ H% x# O- i1 V
( B3 d. Z& b' X2 Y: {/ Y5 ]& uBesser D et al. Expression of nodal, lefty-a, and lefty-B in undifferentiated human embryonic stem cells requires activation of Smad2/3. J Biol Chem 2004;279:45076¨C45084.! B  s  _# w" g( R0 i7 `" [& n

0 F9 Y; r& M8 A- jKorchynskyi O, ten Dijke P et al. Identification and functional characterization of distinct critically important bone morphogenetic protein-specific response elements in the Id1 promoter. J Biol Chem 2002;277:4883¨C4891.1 f; D( i- b8 G! }- \. `
- _; X' i* a  B) O% l0 ~( f
Ho J, de Guise C, Kim C et al. Activin induces hepatocyte cell growth arrest through induction of the cyclin-dependent kinase inhibitor p15INK4B and Sp1. Cell Signal 2004;16:693¨C701.1 f( \9 a  t* j$ _' o9 N/ k, I

; O* v2 c, o+ e8 K( mChambers I, Colby D, Robertson M et al. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 2003;113:643¨C655.1 Q$ W, ]6 I" y( N9 R2 s
( O- Y! ?5 ^/ ?5 Y  _
Mitsui K, Tokuzawa Y, Itoh H et al. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 2003;113:631¨C642.
3 Y: P% r6 j, S( {, s9 A, b7 T  j+ \. B( M3 A
Boyer LA, Lee TI, Cole MF et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005;122:947¨C956.# P( h6 ]$ E& t8 Y

) Y% j5 b8 G9 k2 X8 l* _Brandenberger R, Wei H, Zhang S et al. Transcriptome characterization elucidates signaling networks that control human ES cell growth and differentiation. Nat Biotechnol 2004;22:707¨C716.# Q& u: W/ s9 M, a% j& G! P

6 h7 s% q1 ?6 b1 kSato N, Sanjuan IM, Heke M et al. Molecular signature of human embryonic stem cells and its comparison with the mouse. Dev Biol 2003;260:404¨C413.
/ q# e5 l$ r: C3 A) m: K' O0 v- q7 P1 R8 X: G% q+ a
Babaie Y, Herwig R, Greber B et al. Analysis of Oct4-dependent transcriptional networks regulating self-renewal and pluripotency in human embryonic stem cells. STEM CELLS 2007;25:500¨C510.
! G6 l, Q$ X( g9 ^2 ^6 C9 m' ]4 E& t9 ^% u$ W
Wang L, Li L, Menendez P et al. Human embryonic stem cells maintained in the absence of mouse embryonic fibroblasts or conditioned media are capable of hematopoietic development. Blood 2005;105:4598¨C4603.
" _& V: p3 M: Y7 F& a  d: B. m# u0 B; [6 |. w
Levenstein ME, Ludwig TE, Xu RH et al. Basic fibroblast growth factor support of human embryonic stem cell self-renewal. STEM CELLS 2006;24:568¨C574.
3 K1 ]. I; R7 |9 s4 v: e! S; _. j1 }$ |: J6 ?
Suzuki A, Raya A, Kawakami Y et al. Nanog binds to Smad1 and blocks bone morphogenetic protein-induced differentiation of embryonic stem cells. Proc Natl Acad Sci U S A 2006;.

Rank: 2

积分
166 
威望
166  
包包
1997  
沙发
发表于 2015-5-27 17:09 |只看该作者
谢谢哦  

Rank: 2

积分
75 
威望
75  
包包
2118  
藤椅
发表于 2015-6-26 18:01 |只看该作者
真好。。。。。。。。。  

Rank: 2

积分
161 
威望
161  
包包
1862  
板凳
发表于 2015-7-14 14:55 |只看该作者
干细胞之家微信公众号
留个脚印```````  

Rank: 2

积分
80 
威望
80  
包包
1719  
报纸
发表于 2015-7-27 11:35 |只看该作者
长时间没来看了 ~~  

Rank: 2

积分
107 
威望
107  
包包
1889  
地板
发表于 2015-8-3 11:43 |只看该作者
做一个,做好了,请看  

Rank: 2

积分
107 
威望
107  
包包
1889  
7
发表于 2015-8-19 11:58 |只看该作者
看完了这么强的文章,我想说点什么,但是又不知道说什么好,想来想去只想  

Rank: 2

积分
79 
威望
79  
包包
1769  
8
发表于 2015-8-21 06:23 |只看该作者
嘿嘿......哈哈......呵呵.....哟~呼  

Rank: 2

积分
84 
威望
84  
包包
1877  
9
发表于 2015-8-26 19:42 |只看该作者
人气还要再提高  

Rank: 2

积分
162 
威望
162  
包包
1746  
10
发表于 2015-9-11 06:43 |只看该作者
干细胞研究还要面向临床
‹ 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, 2024-5-14 05:15

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.