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: 391495|回复: 231
go

Absence of Suppressor of Cytokine Signalling 3 Reduces Self-Renewal and Promotes [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:09 |只看该作者 |倒序浏览 |打印
作者:Ariel Forraia, Kristy Boylea, Adam H. Harta, Lynne Hartleya, Steven Rakarb, Tracy A. Willsona, Ken M. Simpsona, Andrew W. Robertsa, Warren S. Alexandera, Anne K. Vossa, Lorraine Robba作者单位:a The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia;
: Q0 F9 a& w5 g1 S1 Z- q1 ?                  
9 Y" u- C/ `5 j4 H) ?                  ) X5 a+ S9 l# f) f/ o" w. l
          ! y5 b8 N, j% |2 w" ?7 F/ ?
                         - S. B; A- [0 V  s2 L1 j1 a
            2 a9 q: o1 G* |) t# T8 |
            
! y( N# q2 u+ z0 F, O1 E            
7 n, z0 F' C- u9 [0 Y8 o+ d            " U- y% W# _3 _+ {. }
                     
# {/ y# f7 I2 e7 \) k# `; m% i9 K7 R        
) \* f$ K* h( v" s        0 d1 E' s4 d1 D4 v7 q% i8 U0 E; w
        
; y8 x8 T8 }0 Q3 e7 k4 a$ p          【摘要】: D8 A/ y2 ~0 a" M. D" ^
      Leukemia inhibitory factor (LIF) is required to maintain pluripotency and permit self-renewal of murine embryonic stem (ES) cells. LIF binds to a receptor complex of LIFR-ß and gp130 and signals via the Janus kinase¨Csignal transducer and activator of transcription (JAK¨CSTAT) pathway, with signalling attenuated by suppressor of cytokine signalling (SOCS) proteins. Recent in vivo studies have highlighted the role of SOCS-3 in the negative regulation of signalling via gp130. To determine the role of SOCS-3 in ES cell biology, SOCS-3¨Cnull ES cell lines were generated. When cultured in LIF levels that sustain self-renewal of wild-type cells, SOCS-3¨Cnull ES cell lines exhibited less self-renewal and greater differentiation into primitive endoderm. The absence of SOCS-3 enhanced JAK¨CSTAT and extracellular signal¨Crelated kinase 1/2 (ERK-1/2)¨Cmitogen-activated protein kinase (MAPK) signal transduction via gp130, with higher levels of phosphorylated STAT-1, STAT-3, SH-2 domain¨Ccontaining cytoplasmic protein tyrosine phosphatase 2 (SHP-2), and ERK-1/2 in steady state and in response to LIF stimulation. Attenuation of ERK signalling by the addition of MAPK/ERK kinase (MEK) inhibitors to SOCS-3¨Cnull ES cell cultures rescued the differentiation phenotype, but did not restore proliferation to wild-type levels. In summary, SOCS-3 plays a crucial role in the regulation of the LIF signalling pathway in murine ES cells. Its absence perturbs the balance between activation of the JAK¨CSTAT and SHP-2¨CERK-1/2¨CMAPK pathways, resulting in less self-renewal and a greater potential for differentiation into the primitive endoderm lineage.
: k1 r* u! ^# \' |          【关键词】 SOCS- Embryonic stem cells Endoderm
4 Q) M: r. Z# X- G( H& [0 M! c                  INTRODUCTION
; b. w1 _8 s# T
+ V5 |' y# K; A+ V- r# M/ LPropagation of pluripotent murine embryonic stem (ES) cells is maintained by the cytokine leukemia inhibitory factor (LIF) .
( E2 h6 L. @) R' I9 U( q4 S6 S3 W( o0 k3 G8 I6 j. H
In ES cells, activation of the Ras¨CERK-1/2¨CMAPK signalling pathway via gp130 is dependent on phosphorylation of the SH2 domain¨Ccontaining cytoplasmic protein tyrosine phosphatase 2 (SHP-2). Phosphorylation of a single tyrosine residue (Y757) in murine gp130 is necessary and sufficient for recruitment of SHP-2, leading to its tyrosine phosphorylation in a JAK-1¨Cdependent manner .
  e2 a# h( x, j* z" Q8 p; F
% I& u8 ~9 r; v) T6 @$ sAn increasing number of studies has shown that the suppressor of cytokine signalling 3 (SOCS-3) protein is a key regulator of signalling mediated via gp130. SOCS proteins contain a central SH2 domain and a carboxy-terminal SOCS box. Each of these domains is thought to have a separate function in regulating cytokine signalling. The SH2 domain interacts with phosphorylated tyrosine residues in tyrosine kinases and cytokine receptors and can negatively regulate their activity .6 c3 g- [6 ^$ p' X
2 q; t* E3 A) V$ l, L- q* `
Gene-targeting experiments have reinforced the notion that SOCS-3 is a major physiological regulator of signalling via gp130. SOCS-3¨Cnull embryos die at midgestation as a result of placental failure, and this can be rescued by a reduction of signalling via LIFR-ß . In order to understand the role of SOCS-3 in ES cells, we derived SOCS-3¨Cnull ES cell lines. In standard culture conditions, SOCS-3¨Cnull ES cell lines show less proliferation and greater differentiation to endoderm. In steady state, STAT-3 phosphorylation is greater in SOCS-3¨Cnull ES cells and, in contrast to WT cells, SHP-2 is phosphorylated. In response to LIF stimulation, STAT-3, SHP-2, and ERK-1/2 activation are prolonged in SOCS-3¨Cnull ES cells. Overall, we demonstrate that SOCS-3 plays a crucial role in regulating LIF signalling in ES cells and that its absence results in alterations in self-renewal and differentiation.
- M: m' ~- x- Z) F+ ~* _
- {) I% I; M6 o' Y" KMATERIALS AND METHODS4 [. [. L  W; I& w

5 Q) r+ ]; [, |  g8 u3 R6 ^) T5 o( ^' _Generation of SOCS-3¨CNull Mice and ES Cell Lines
3 ?7 \3 x5 N! ~: M% ]& d& L& m# e  B! W: d* H1 D5 r9 u
The SOCS-3 gene-targeting vector has been described previously . The genotype of the ES cell lines was determined by Southern blotting of BamHI-digested DNA with 5' and 3' genomic DNA probes.7 O7 l4 x/ l3 R8 ]

: T# e7 k/ r9 n& jCell Culture
4 r" s* M. F+ j) m8 K# C* O1 w
' r  I$ b/ V9 y" EES cells were maintained on a layer of irradiated primary mouse embryonic fibroblasts in ES medium¡ªDulbecco¡¯s modified Eagle¡¯s medium (DMEM) supplemented with 4.5 g/l glucose, 3.4 g/l NaHCO3, 15% (vol/vol) batch-tested fetal calf serum (FCS) (Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com), 0.1 mM 2-mercaptoethanol (Sigma), and 0.1 mM nonessential amino acids (Invitrogen, Carlsbad, CA, http://www.invitrogen.com)¡ªas described elsewhere . To test the effect of MEK inhibitors, ES cells were plated at 0.5 x 106 cells per well of a six-well tissue culture plate in standard ES cell culture medium and were supplemented with either 10 µM UO126 (Promega, Madison, WI, http://www.promega.com), 50 µM PD98059 (Promega), or 1 µl/ml dimethylsulfoxide (DMSO). The medium was changed every 2 days.
- a& R0 `) S8 W. a1 u2 Z
7 Q! k; ]; g) I0 Z1 SReverse Transcription-Polymerase Chain Reaction
2 u7 T0 i- H4 n( K* D" B% j8 K% M2 D0 ^& t' f2 y
Total RNA was extracted from feeder-independent ES cell lines using the RNeasy Mini RNA kit (Qiagen, Valencia, CA, http://www1.qiagen.com). DNaseI-treated samples were reverse transcribed (RT) using Superscript III (Invitrogen), the resultant cDNA preparations were standardized, and polymerase chain reaction (PCR) was performed as described elsewhere . Primers for Socs3 were: 5'-AGATTTCGCTTCGGGACTAGC-3'5'-CTGGGTCTTGACG-CTCAAGCT-3'.
( D. H4 p/ B5 i. A* c1 B( J" [1 w0 j! g6 Z
Indirect Immunofluorescence
% F6 l" E( p1 U7 C8 ~, ^3 _
7 s) h' j% t9 x! t( h' USOCS-3¨Cnull and WT ES cells were plated onto gelatinized potassium hydroxide (KOH)-treated glass coverslips at a density of 1 x 105 cells per coverslip. Cells were cultured for 48 hours, washed with phosphate-buffered saline (PBS), and fixed in 4% paraformaldehyde (PFA) for 10 minutes at room temperature. Immunofluorescent staining was performed as previously described  using antibodies specific for Oct4 and GATA-4 (Santa Cruz Biotechnology Inc., Santa Cruz, CA, http://www.scbt.com). After application of the secondary antibody (Alexa Fluor 594 goat anti-mouse IgG, Molecular Probes Inc., Eugene, OR, http://probes.invitrogen.com), the cells were washed, counterstained with 4',6-diamidino-2-phenylindole (DAPI) mounted in DAKO (Glostrup, Denmark, http://www.dako.com) mounting media and viewed with a Zeiss Axioplan 2 (Carl Zeiss, Jena, Germany, http://www.zeiss.com) microscope. Images were captured with a Zeiss Axiocam (Carl Zeiss) and processed with Axiovision software (Carl Zeiss).
2 e# a1 j' g0 I  q+ Y
# |8 I! F' T! f: f% O* mMicroarray Analysis5 F$ C5 Q% K) }$ {

- M5 t% k" j( T! a# F- f4 QTotal RNA was extracted from two independent WT and SOCS-3¨Cnull ES cell lines grown on primary mouse embryonic fibro-blasts in standard culture conditions using Qiagen RNeasy Mini RNA purification columns. The RNA from each cell line was quantitated using an Agilent 2100 Bioanalyser (Agilent Technologies, Palo Alto, CA, http://www.agilent.com). cDNA was synthesized from 5 µg of RNA according to Affymetrix methodology, and biotin-labeled cRNA was synthesized using the Affymetrix Enzo BioArray High Yield RNA Transcript Labeling Kit (Affymetrix, Santa Clara, CA, http://www.affymetrix.com). Affymetrix mouse genome (MOE 430 2.0, Affymetrix) GeneChips were hybridized with 10 µg of biotin-labeled cRNA according to the manufacturer¡¯s protocol. Expression levels were calculated using Robust Multichip Analysis (RMA) . We selected, as differentially expressed, those genes with an absolute value of t* = 5. This threshold was derived by examining a plot of the quantiles of t* against the quantiles of the t-distribution on the appropriate degrees of freedom, and looking for departures from linearity. All calculations were done in R, using the "affy" and "limma" packages. The microarray data contained in this manuscript have been submitted to the GEO database, available at http://ncbi.nlm.nih.gov/geo.
" m$ L: S' k9 F
6 Z9 C+ a; J# x8 R/ Q. ?  xChimera Generation, Induction of Teratomas, and Tumor Histology6 p' j  f3 F/ i' d

& ]0 O( c' H) u" T# H6 QChimeric embryos were generated by injection of ES cells into blastocysts obtained from intercrosses of mice carrying the ROSA-26 lacZ gene trap . To generate teratomas, 4-week-old nude mice were injected s.c. in opposing flanks with 1 x 106 WT or SOCS-3¨Cnull ES cells in 100 µl of PBS, and tumors were harvested 4¨C6 weeks later. Tissue was fixed in Bouin¡¯s fixative, dehydrated, embedded in paraffin, sectioned at 0.4 µm, and stained with hematoxylin and eosin. Eight WT and eight SOCS-3¨Cnull tumors from four ES cell lines were examined. Animal studies were approved by the Melbourne Health Research Directorate Animal Ethics Committee.% f1 a/ I; k$ a( z4 k
- b! N  _# Z& b! _# F
Immunoprecipitation and Western Blotting
) L0 ]8 G" L# S0 t- i
" K. e4 V2 B/ _For cytokine stimulation experiments, feeder-independent ES cell lines were grown to near confluence in standard ES cell medium and then washed three times in PBS and cultured in ES cell medium with 0.5% FCS and without LIF for 4 hours prior to restimulation with LIF (1000 units/ml). For immunoprecipitation, cells were lysed in KALB lysis buffer (1% Triton X-100(vol/vol), 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA) supplemented with 1 mM Na3VO4, 1 mM NaF, 1 mM phenylmethylsulfonyl fluoride (PMSF), and complete protease inhibitor mixture (Roche Applied Science, Indianapolis, https://www.roche-applied-science.com). Total cell lysates were pre-cleared with 50% slurry protein A sepharose (PAS), incubated with 2 µg SH-PTP2 antibody (c-18, Santa Cruz), and immunoprecipitated with PAS. For Western blotting, near confluent cells were lysed in RIPA lysis buffer (1% Triton X-100(vol/vol), 0.1% SDS(w/vol), 1% sodium deoxycholate, 150 mM NaCl, 20 mM Tris-HCl pH 7.5, 0.01% sodium azide) supplemented as above. Lysates or immunoprecipitates were subjected to SDS-PAGE separation and immunoblotting using antibodies specific for SOCS-3 (Immuno-Biological Laboratories Co., Ltd., Gunma, Japan, http://www.ibl-japan.co.jp), STAT-3 (Santa Cruz), STAT-1 (BD Signal Transduction, Franklin Lakes, NJ, http://www.bdbiosciences.com), and ERK-1/2 (Cell Signalling Technology, Beverly, MA, http://www.cellsignal.com) and antibodies specific for the phosphorylated forms of STAT-3, STAT-1, ERK-1, ERK-2, and SHP-2 (Cell Signalling). The densitometry analysis was performed on scanned Hyperfilm ECL autoradiographs using a Molecular Dynamics Densitometer and ImageQuant software (GE Healthcare Life Sciences, Piscataway, NJ, http://www.amersham.com).: Q2 |; O& I! w8 }$ }1 z5 I  e
: U& `: R- E- u0 Z7 C
Online Supplemental Material" I5 R! T! u/ U6 q) S8 O9 y( \" G

/ E9 A2 {+ q  L% h6 e4 }2 k* l" jSupplemental online Figure 1 shows the generation and validation of SOCS-3¨Cnull ES cell lines. Supplemental online Figure 2 shows alterations in global gene expression in SOCS-3¨Cnull ES cells, using an MA plot. Supplemental online Tables 1 and 2 list genes with fivefold or greater altered expression in SOCS-3¨Cnull ES cell lines.6 y3 ^' {* q) ^" b3 j% B7 v& m$ ~

  Z7 h" T5 M/ R: Z4 T* nRESULTS
2 b0 J9 H* |- Z0 d6 l# S: G- _$ c, v
8 G+ s  b5 x) ?Generation of SOCS-3¨CNull ES Cell Lines5 }2 o, R  B9 h9 y4 q

% t$ E4 J3 }, b! K% X1 S, g% ZTo explore the function of SOCS-3 in ES cells, we initially attempted to generate SOCS-3¨Cnull ES cell lines by serially inactivating each allele using targeting constructs with selection cassettes for neomycin resistance or hygromycin resistance . From 96 blastocysts, 55 ES cell lines were obtained, of which 12 were homozygous for targeted deletion of Socs3. RT-PCR and immunoblotting demonstrated that SOCS-3 RNA and protein were not detectable in the SOCS-3¨Cnull ES cell lines (supplemental online Fig. 1). In all experiments reported here, WT ES cell lines outgrown from blastocysts in the same experiment as the null lines were used as controls, and all results shown are representative of 3¨C6 WT and SOCS-3¨Cnull ES cell lines.
0 ]" p' C* y0 o' G' }6 W! G6 L
SOCS-3¨CNull ES Cell Lines Exhibit Reduced Proliferation and Self-Renewal. k+ @/ _( M7 a9 |$ ?& H, V& }! [
. S4 b5 @) k! i& }
SOCS-3¨Cnull ES cell lines could be continuously passaged in culture and were recoverable after cryostorage. Initially, all lines were maintained on a feeder layer of primary mouse embryonic fibroblasts. To facilitate biochemical analysis, feeder-independent lines were established. During routine culture, the feeder-dependent and feeder-independent SOCS-3¨Cnull ES cell lines were noted to grow more slowly than the WT lines, but could be serially passaged for at least 3 months. In SOCS-3¨Cnull ES cell cultures, around 50% of the colonies were observed to be partially differentiated, with cells at the periphery of the individual colonies adopting a flattened, refractile morphology. (Fig. 1A¨C1H). To quantitate the proliferative defect in the null lines, viable single cells were plated at low density (100 cells per well in 24-well tissue culture plates) and the number of cells per well was counted daily. In comparison with WT ES cells, the SOCS-3¨Cnull cells demonstrated markedly lower proliferation (Fig. 1I). To assess self-renewal, the number of colonies present at day 3 after plating was ascertained and the colonies were scored as undifferentiated, partially differentiated, or fully differentiated. As shown in Figure 1J, clonogenicity of the null lines was markedly lower and, in addition, SOCS-3¨Cnull ES cells gave rise to a higher proportion of colonies containing differentiated cells. To assess LIF responsiveness in the null lines, cells were plated in different concentrations of LIF, and colony morphology was scored daily. At 10 units of LIF, both WT and SOCS-3¨Cnull ES cell cultures contained
0 L, M; _9 v% U8 U4 G" o
6 b; Y( X. g& x' W/ i$ B$ CFigure 1. SOCS-3¨Cnull ES cells exhibit altered morphology, proliferation, and clonogenicity. (A¨CH): Morphology of two independent wild-type (WT) and SOCS-3¨Cnull ES cell lines, grown in standard ES cell culture conditions (15% fetal calf serum and 1000 units/ml LIF), on (A¨CD) and after weaning off (E¨CH) PMEFs. Note the partially differentiated appearance of the colonies arising from the null lines. Differentiated cells present in SOCS-3¨Cnull ES cell lines cultured on or off PMEFs are indicated by arrows. ES indicates undifferentiated ES cell colonies. (I): Growth of WT and SOCS-3¨Cnull ES cell lines. At each time point, the mean and standard deviation for triplicate wells is shown. Similar results were obtained with four WT and four SOCS-3¨Cnull lines. (J): Number of colonies present at day 3 after plating 100 cells in standard ES cell culture conditions. Colonies were fixed, stained, and scored as undifferentiated, mixed, or differentiated. Means and standard deviations for triplicate wells are shown. (K): WT and SOCS-3¨Cnull colonies were enumerated as for (J) after 3 days of culture in standard ES cell medium with concentrations of LIF as shown. Abbreviations: SOCS-3, suppressor of cytokine signalling 3; ES, embryonic stem cell; PMEF, primary mouse embryonic fibroblasts; W and WT, wild type; N, SOCS-3 null; LIF, leukemia inhibitory factor. Results are shown for a single representative experiment. Scale bars = 25 µm.6 }  Q# P7 N: b! w3 `! s4 g

5 d. O4 g! m/ v& oSOCS-3¨CNull ES Cells Differentiate into Endoderm in the Presence of LIF' N+ S$ j3 i. F9 @8 K
: P/ [5 h5 U  y& P: b
When murine ES cells are propagated in standard conditions with FCS and LIF, minimal differentiation is observed. In cultures of SOCS-3¨Cnull ES cell lines, however, we observed a greater proportion of partially differentiated ES cell colonies, together with dispersed, refractile cells with stellate morphology, reminiscent of primitive endoderm cells . To establish the identity of the differentiated cell types we used RT-PCR to examine the expression of genes associated with the undifferentiated state or with differentiation into different cell lineages. In keeping with the observation that SOCS-3¨Cnull ES cells could be cultured continuously and formed colonies containing cells with typical ES cell morphology, the pluripotential cell markers Nanog, Oct4, and Zfp42 were expressed. Markers of mesoderm (Brachyury, Mixl1), neuroectoderm (Isl1, Sox1, Otx2), and trophectoderm (Hand1) were not detected in the SOCS-3¨Cnull ES cell lines. In contrast, there was a striking upregulation of endoderm-specific gene expression. Endodermal transcription factors Tcf2, Gata4, Gata6, Hnf4a, and COUPTF1 were upregulated. Markers of parietal endoderm, Lamb1¨C1 and Dab2, were expressed, but visceral endoderm markers, Afp and Ttr, were not, suggesting that the differentiated cells arising in the SOCS-3¨Cnull ES cell cultures were of the primitive endoderm type (Fig. 2A). Immunohistochemical analysis detected Oct4 protein in WT and SOCS-3¨Cnull ES cells (Fig. 2B¨C2D). GATA-4 protein was not detectable in WT ES cells, but in SOCS-3¨Cnull cultures was detectable in cells at the edge of partially differentiated colonies (Fig. 2E¨C2G). GATA-4, but not Oct4, was detectable in the dispersed cells present in the SOCS-3¨Cnull ES cell cultures, thereby establishing their identity as endoderm. (Fig. 2H¨C2J).  z5 b& o( _! T4 y" Q7 S/ T: R

6 V9 V% ^+ }0 u6 U" R- ]2 WFigure 2. In the absence of SOCS-3, ES cells undergo differentiation to primitive endoderm. (A): Reverse transcription-polymerase chain reaction analysis demonstrates that SOCS-3¨Cnull ES cells express typical stem cell markers and show increased expression of primitive endoderm markers. (B¨CG): Indirect immunofluorescence was utilized to detect Oct4 (red) and GATA4 (green) in wild-type and SOCS-3¨Cnull ES cell cultures. In merged images (H¨CJ) and (B, E, H) Oct-4 but not GATA-4, is detectable in WT ES cell colonies. (C, F, I): In a partially differentiated SOCS-3¨Cnull ES cell colony, Oct-4 is readily detectable and GATA4 is seen in cells at the periphery of the colony. (D, G, J): Undifferentiated Oct-4 positive SOCS-3¨Cnull ES cell colony and dispersed, differentiated cells with typical endodermal morphology (arrows) in which GATA-4 protein is detected. Nuclei of all cells in (B¨CG) are stained with 4',6-diamidino-2-phenylindole. Abbreviations: SOCS3, suppressor of cytokine signalling 3; ES, embryonic stem cell;  ve, positive control (embryoid body or embryonic day 9 cDNA); ¨Cve, negative (no cDNA) control; WT, wild-type. Scale bars = 50 µm., e4 E" R8 J% m* |+ S5 X' k; H

& D1 T, y" h7 d' TTo extend the analysis of transcriptional differences between SOCS-3¨Cnull and WT ES cells, gene expression in WT and SOCS-3¨Cnull ES cell lines grown in standard ES cell culture medium on a feeder layer was compared. The gene expression levels of 45,000 known and predicted gene transcripts present on the Affymetrix MOE430 GeneChip were assayed, and expression of a large number of genes was significantly different in the null lines (supplemental online Fig. 2). The 161 genes upregulated and 61 genes downregulated more than fivefold in the SOCS-3¨Cnull ES cell lines are listed in supplemental online Table 1 and Table 2. As indicated by the RT-PCR results, early endoderm-specific genes were highly upregulated in the SOCS-3¨Cnull ES cell lines, but genes characteristically expressed in embryonic definitive endoderm (e.g., Afp, Serpini1, Alb1, Ipf1) were not upregulated, further demonstrating that a major consequence of the absence of SOCS-3 is the induction of a proportion of ES cells to adopt a primitive endodermal fate (supplemental online Table 1). Of the 161 upregulated genes, 25% had previously been documented to be expressed in endoderm of the preimplantation embryo, or during differentiation of embryoid bodies or embryocarcinoma cell lines to form endoderm . The expression of genes associated with trophectoderm was similar in WT and mutant cell lines, and genes marking the neuroectodermal and mesodermal lineages were not upregulated. Expression of genes associated with pluripotency, including Nanog, Oct4, Zpf42, and Foxd3, was not significantly different in the WT and SOCS-3¨Cnull ES cell lines. None of the other seven members of the SOCS gene family showed altered gene expression in the SOCS-3¨Cnull ES cells, indicating that functional compensation by other SOCS genes was unlikely to be contributing to the observed phenotype (supplemental online Table 2; data not shown).$ @& w( L4 a4 [6 q" P7 S3 q4 Q. X. C
' r) q) N" N9 O) h7 _
SOCS-3¨CNull ES Cell Lines Can Differentiate into Cells Derived from All Three Germ Layers4 T0 c  b" K2 s9 s

5 e& ~, M* A0 v- z* y2 m$ w( t/ zTo determine whether the absence of SOCS-3 affects ES cell differentiation potential, expression of cell lineage¨Cspecific marker genes was assessed in WT and SOCS-3¨Cnull ES cell cultures after LIF withdrawal. In addition, the lines were cultured at low density in differentiation medium to allow formation of embryoid bodies (Fig. 3A). Embryoid bodies formed at 5- to 10-fold lower frequencies from SOCS-3¨Cnull ES cell lines (data not shown). RNA was prepared from adherent cultures 5 days after LIF withdrawal and from embryoid bodies at day 6, and RT-PCR was performed. Differentiated WT and SOCS-3¨Cnull ES cell cultures and embryoid bodies expressed markers of mesoderm, ectoderm, and endoderm (Fig. 3B). To evaluate the differentiation potential of the SOCS-3¨Cnull ES cell lines in vivo, we injected ES cells s.c. into nude mice to induce the formation of teratomas. These benign tumors contain well differentiated tissues of ectodermal, mesodermal, and endodermal origin . Differentiation profiles of the resulting teratomas were assessed by histological examination. Both WT and SOCS-3¨Cnull teratomas had a heterogeneous differentiation profile, with cells of ectodermal lineage (neuronal structures), mesodermal lineage (smooth muscle, cartilage), and endodermal lineage (goblet cells, respiratory epithelium) (Fig. 3C¨C3G). Both WT and SOCS-3¨Cnull ES cell¨Cderived teratomas also contained trophoblast cells (Fig. 3H). When injected into genetically marked blastocysts, the SOCS-3¨Cnull ES cells contributed to all tissues of chimera embryos (data not shown). Together, the results indicate that the absence of SOCS-3 does not affect the capacity of ES cells to differentiate into tissues derived from all three germ layers.7 P5 E, H8 y$ O$ t( ]

  i# A3 V3 Q0 c( o, f6 m5 k6 _Figure 3. SOCS-3¨Cnull ES cells retain the capacity to differentiate into cells derived from all three germ layers. (A): WT and SOCS-3¨Cnull embryoid bodies. (B): Reverse transcription-polymerase chain reaction analysis showing gene expression in WT and SOCS-3¨Cnull ES cells maintained in LIF ( LIF) and after LIF withdrawal (¨CLIF) and in day 6 embryoid bodies (D6 EB). (C¨CH): SOCS-3¨Cnull ES cells were injected s.c. into nude mice, and the resulting teratomas were fixed, sectioned, and stained with hematoxylin and eosin. Tissues derived from neuroectoderm (C), mesoderm (E, F), and endoderm (G) were present. (C): Primitive neuroepithelial tubes. (D): Cartilage. (E): Smooth muscle. (F): Ciliated epithelium. (G): Gut epithelium. (H): SOCS-3¨Cnull embryonic stem cells and WT ES cells (not shown) also gave rise to trophoblast tissue. Abbreviations: SOCS-3, suppressor of cytokine signalling 3; WT, Wild-type; LIF, Leukemia inhibitory factor; SOCS-3¨Cnull ES cells maintained in LIF; D6 EB, in day 6 embryoid bodies; ne, neuroepithelium; c, cartilage; ce, ciliated epithelium; ge, gut epithelium; tgc, trophoblast giant cells. Magnification: x 200.- x9 L  x6 R/ N; C6 r. N
5 W7 ~1 {$ [& p2 x" ?% b
Greater STAT-1, STAT-3, and SHP-2 Activation in the Absence of SOCS-3
9 n/ s9 e$ t: i0 e
, G! \+ F! H+ x8 {! }* o! ZWestern blots were performed using lysates prepared from feeder-independent WT and null cell lines cultured in ES cell medium with LIF (steady state) and from cultures that had been cytokine and serum starved for 4 hours prior to readdition of LIF (1000 units/ml) (Fig. 4). SOCS-3 was present in WT ES cells in steady state and was detected at 30 minutes after LIF stimulation. Steady-state levels of phosphorylated STAT-1 and STAT-3 were higher in the SOCS-3¨Cnull ES cell lines (Fig. 4). After starvation and LIF addition, STAT-3 phosphorylation in WT cells was maximal between 5 and 30 minutes, but by 1 hour had returned to the level seen during routine culture. In contrast, SOCS-3¨Cnull ES cells showed a blunted response to readdition of LIF, exhibiting sustained STAT-3 phosphorylation. After LIF stimulation, ongoing STAT-1 phosphorylation was observed in the SOCS-3¨Cnull ES cells.
) w/ b+ P+ _: c" r. x; e9 r0 E* g1 q" E- F( g( V: W+ |! S9 Y6 p8 X
Figure 4. LIF signalling is deregulated SOCS-3¨Cnull ES cells. Feeder-independent cell lines were grown to near confluence over 48 hours in standard ES cell culture medium, after which they were washed and placed into embryonic stem cell medium containing 0.5% serum without LIF for 4 hours. After starvation, 1000 units/ml LIF was added, and cell lysates were prepared at the indicated times thereafter. Lysates were separated by SDS-PAGE, blotted, and probed with antibodies specific to SOCS-3 or to pSTAT-3, pSTAT-1, or pERK-1/2. Membranes were stripped and reprobed with antibodies to total STAT-3, STAT-1, and ERK-1/2. Total SH2 domain¨Ccontaining SHP-2 was immunoprecipitated from cell lysates, separated by SDS-PAGE, and probed with antibodies specific to phosphorylated SHP-2 (pSHP-2) or total SHP2.  LIF indicates cells maintained in standard culture conditions throughout, ¨CLIF indicates cells harvested after 4 hours of starvation without restimulation. Histograms show densitometric quantitation of mean and standard deviation of the signal from three Western blots. The phosphorylation level is reported as a percentage relative to total STAT-1, STAT-3, SHP-2, or ERK-1/2 protein. Abbreviations: LIF, Leukemia inhibitory factor; SOCS-3, suppressor of cytokine signalling 3; pSTAT, phosphorylated signal transducer and activator of transcription; pERK-1/2, phosphorylated forms of extracellular signal¨Crelated kinase 1/2; ERK; extracellular signal¨Cregulated kinase; SHP-2, cytoplasmic SH2-containing protein tyrosine phosphatase 2; WT, wild type.9 S; r& O/ t- n

2 k  L/ a7 `, VSHP-2 was immunoprecipitated from cellular extracts of WT and SOCS-3¨Cnull ES cells, and SHP-2 phosphorylation was analyzed by Western blotting. Strikingly, in steady-state culture conditions, SHP-2 phosphorylation was detectable in SOCS-3¨Cnull ES cells but not in WT cells. LIF stimulation of WT cells induced SHP-2 phosphorylation that was rapidly attenuated, whereas in SOCS-3¨Cnull ES cells, there was a sustained induction of SHP-2 phosphorylation. The absence of SOCS-3 in ES cells also resulted in greater steady-state MAPK activation. Phosphorylated ERK-1 and ERK-2 proteins were present in greater amounts in steady-state SOCS-3¨Cnull ES cells, and in LIF readdition experiments, phosphorylation of ERK-1/2 lasted longer than with WT cells (Fig. 4). In keeping with previous observations, ERK-2 was the predominant phosphorylated ERK detected in ES cells.
% r& n+ p1 o+ W% e" X$ i; }
. X! d! q; F6 lAddition of Ras¨CMAPK Inhibitors Rescues the SOCS-3¨CNull ES Cell Differentiation Phenotype
  ~2 _3 p/ U1 c8 X" v; {
$ k9 p6 v1 `. r0 N' W+ a! F3 H, ?SHP-2¨CRas¨CERK-1/2¨CMAPK signalling provides a differentiative signal in ES cells. To establish if the upregulation of this pathway in SOCS-3¨Cnull ES cells was responsible for their phenotype, WT and null ES cell lines were treated with the MEK inhibitors PD98059 or U0126 . Feeder-independent WT and null ES cells were cultured in the presence of inhibitor or carrier (DMSO) and assessed for the presence of differentiating colonies. The SOCS-3¨Cnull ES cell differentiation phenotype was completely reversed by the addition of either inhibitor to the culture medium (Fig. 5A¨C5H). This was confirmed by RT-PCR analysis, which showed that treatment with MEK inhibitors reduced Gata4 expression in the SOCS-3¨Cnull lines to a level similar to that observed in WT cells (Fig. 5I). Immunoblotting of lysates from treated cells showed less ERK-1/2 phosphorylation in the SOCS-3¨Cnull ES cells after treatment with inhibitor (Fig. 5J). To assess whether MEK inhibition affected proliferation and clonogenicity, PD98059- or U0126-treated WT and SOCS-3¨Cnull cells were plated at low density; the number of cells per well was counted daily and the colony number and morphology on day 3 after plating was scored (Fig. 5K, 5L). Addition of U0126 or PD98059 (not shown) reduced proliferation and clonogenicity in the WT cell lines. This effect was observed even at the lowest concentration of either inhibitor that was sufficient to inhibit ERK-/2 phosphorylation (not shown). In contrast to the effect on the differentiation phenotype, the lower proliferative capacity and clonogenicity of the SOCS-3¨Cnull lines was not rescued by inhibition of Ras¨CERK-1/2¨CMAPK signalling." D* D: [; ?6 g

% P' [: c  L4 YFigure 5. Treatment of SOCS-3¨Cnull embryonic stem (ES) cells with mitogen-activated protein kinase/ERK kinase (MEK) inhibitors prevents differentiation into endoderm. (A¨CH): Feeder-independent wild-type (WT) and SOCS-3¨Cnull ES cell lines were trypsinized and replated in ES cell medium (¨C) with or without the MEK inhibitors PD98059 or U0126 or the carrier DMSO. (I): Reverse transcription-polymerase chain reaction analysis of gene expression in WT and SOCS-3¨Cnull lines after treatment with U0126 shows that Gata4 expression in treated SOCS-3¨Cnull lines, but not in controls, is reduced to near WT levels. (J): Cell lysates prepared from WT and SOCS-3¨Cnull ES cell lines prior to and after treatment with U0126 were separated by SDS-PAGE, blotted, and probed with antibodies specific to phosphorylated forms of ERK-1/2 (pERK-1/2). Membranes were stripped and reprobed with antibodies to total ERK-1/2. Note the difference in the amount of total lysate loaded in the WT and SOCS-3¨Cnull lanes. (K, L): WT and SOCS-3¨Cnull ES cells were treated with MEK inhibitors and then plated at low density. W, wild-type cells; N, SOCS-3¨Cnull ES cells. Cell (K) and colony (L) counts were performed daily to assess proliferation and response to leukemia inhibitory factor (LIF) withdrawal. Data shown in (L) are the mean triplicate wells from a representative experiment. Similar results were obtained with two WT and four SOCS-3¨Cnull ES cell lines. Abbreviations: SOCS-3, suppressor of cytokine signalling 3; DMSO, dimethylsulfoxide; W and WT, wild type; ERK, extracellular signal¨Cregulated kinase; N, SOCS-3 null. Scale bars = 25 µm.! U) Y( Z$ S& P8 M9 |

& P  R: @( Z% F' B- N" U# KDISCUSSION9 _, i. u$ E! i$ t' @, j& v% K
5 n" G. \3 p4 |' H9 [% e
To ascertain the role of SOCS-3 in ES cells we generated SOCS-3¨Cnull ES cell lines. Null cell lines could not be derived by gene targeting but were successfully isolated from preimplantation blastocysts. Our inability to derive doubly targeted clones by gene targeting likely reflects the lower proliferation capacity and clonogenicity of SOCS-3¨Cnull ES cells. SOCS-3¨Cnull ES cell lines exhibited less self-renewal and a greater propensity to differentiate into endoderm. Undifferentiated SOCS-3¨Cnull ES cells expressed stem cell markers and retained the capacity to differentiate into tissues of ectodermal, mesodermal, and endodermal origin in vitro and in vivo. Strikingly, the differentiative phenotype could be rescued by inhibition of MAPK signalling.0 I2 f0 F8 a8 u2 X% F; e7 C

% D' G7 G9 u! b( |The ablation of SOCS-3 resulted in greater intensity and duration of activation of both the JAK¨CSTAT and the Ras¨CERK-1/2¨CMAPK signalling cascades both in the steady state and in response to LIF signalling. The null cells exhibited higher STAT-3 activation in steady-state cultures and sustained activation of STAT-3 after starvation and LIF readdition. Unlike WT cells, STAT-1 was constitutively phosphorylated. Phosphorylated SHP-2 was not detectable in WT ES cells maintained in LIF and serum, but in the absence of SOCS-3, phosphorylated SHP-2 was readily detectable, presumably as a result of a lack of competition from SOCS-3 for binding to Y757 of gp130. After cytokine stimulation, greater and sustained SHP-2 activation was observed in the null cells, whereas in WT cells, SHP-2 phosphorylation waned as SOCS-3 was upregulated. Downstream of SHP-2, ERK-1/2 phosphorylation was greater in the null cells. In other systems, SOCS-3 has been shown to positively regulate MAPK activation. In human T cells, SOCS-3 is tyrosine phosphorylated in response to multiple stimuli and binds to and inactivates Ras/GTPase-activating protein (GAP), leading to Ras¨CERK-1/2¨CMAPK activation . It is not known whether a similar mechanism operates in murine ES cells.7 I. K2 Z9 D6 [+ a5 V
0 l# C7 v6 \  Y# t- \8 w+ ~
STAT and SHP-2¨CRas¨CERK signalling via gp130 are key regulators of cellular homeostasis. In vitro, the simultaneous activation of Ras¨CERK¨CMAPK and STAT-1/3 has been repeatedly shown to generate opposing signals, the balance of which determines the biological outcome and, in vivo, balanced activation of the two signalling cascades is required to prevent disease . Inhibition of ERK-1/2¨CMAPK signalling does not replace the requirement for STAT-3 signalling, but rather works synergistically with it.' d; O/ b: n' j6 d
3 I2 V$ i$ q0 l
In SOCS-3¨Cnull ES cells, constitutive upregulation of Ras¨CERK-1/2¨CMAPK signalling provides a differentiative signal. This is opposed by augmented self-renewal signals via phosphorylated STAT-3. However, increased STAT-3 activation is not sufficient to prevent differentiation when Ras¨CERK-1/2¨CMAPK signalling is activated. We hypothesize that this mixture of self-renewal and differentiative signals drives the phenotype observed in the SOCS-3¨Cnull ES cell cultures and that alterations in the thresholds of self-renewal and differentiative signals in individual cells result in the partial differentiation phenotype that can be reversed by pharmacological inhibition of MEK. During continuous subculturing, the undifferentiated cells in SOCS-3¨Cnull ES cell cultures would be expected to have a preferential replating advantage over the more differentiated cells, thus supporting their long-term dominance .
8 c* \5 y6 R( T/ a, k
( w0 E+ ~) M# O0 {9 J" I9 hES cell proliferation was markedly lower in the absence of SOCS-3 despite high levels of steady-state STAT-3 phosphorylation. The role of STAT-3 activation in ES cell proliferation is unclear. While it is well established that less STAT-3 activation leads to abrogation of ES cell self-renewal, it has not been shown to affect proliferation . The lower proliferation observed in the SOCS-3¨Cnull ES cell lines may be in part a result of dysregulated STAT-3 activation, but it is likely that alterations in other, as yet unidentified, signals also affect proliferation.
( i$ N' E: g! ~4 i/ r# W
" d: v9 i9 j+ g% }9 F( |0 s9 GLIF is only able to sustain ES cells in the presence of serum, suggesting that additional factors are required. Recently, evidence has emerged that bone morphogenetic proteins (BMPs) may act in combination with LIF to sustain self-renewal of murine ES cells by inducing the expression of inhibitors of differentiation (Id) genes . The analysis of the SOCS-3¨Cnull ES cell phenotype reported here was conducted using ES cell media supplemented with FCS. In future experiments, it will be important to dissect the contribution of individual factors to the SOCS-3¨Cnull phenotype.
6 U6 g" L# d+ I6 f
1 q; s2 {: H& ?6 ^+ i& SThe in vivo relevance of the LIF signalling pathway for early embryo development is uncertain. During embryogenesis, the inner cell mass (ICM) exists transiently and does not act as a long-term stem cell compartment. It is not clear whether a population equivalent to the ES cell ever exists in vivo. Neither LIF, LIFR-ß, nor gp130 mutants show defects in the development of the ICM or early epiblast .
2 v- V$ h% R  v; C% f5 ]: J" q4 f! `; [# x% e; o/ u
In summary, activation of cytokine signalling pathways in murine ES cells alters cell fate and potency. Results of SOCS-3 overexpression in ES cells, together with the data presented here on the effects of SOCS-3 ablation in ES cells, point to a key role for SOCS-3 in regulating signals emanating from the LIFR-ߨCgp130 receptor complex in ES cells. In doing so, SOCS-3 regulates ES cell self-renewal and insulates ES cells from the functional consequence of lineage priming via the Ras¨CERK¨CMAPK signalling pathway.
4 j0 R' i  |$ S( m# D& D8 O, }. b; g2 p5 g$ l6 [
DISCLOSURES. f5 i6 }3 J& x* d

9 i# ^# [- ?9 S$ I& S. xThe authors indicate no potential conflicts of interest.
0 U" _* k* X. x8 R# g6 F2 {
+ S# _6 c! o% N$ B! m+ E5 nACKNOWLEDGMENTS
6 t' [! v1 d5 M' \3 b  M) X0 O; Z# k3 L
We thank Lucille Vollaire for karyotyping, Ruili Li for blastocyst injections, and Janelle Lochland for genotyping. We also thank Tim Thomas for genotyping the ES cell lines¡ªderived de novo from SOCS-3 heterozygous intercrosses. We are grateful to Prof. Terry Speed and Dr. Gordon Smyth for discussion regarding analysis of the microarray data and Profs. Nicos Nicola and Doug Hilton for comments on the manuscript. The project was supported by the National Health and Medical Research Council of Australia program grant 257500, NIH grant CA22556 and Zenyth Therapeutics Limited. A.F. and K.B. contributed equally to this work.
1 t" m$ |0 B# u0 Y4 ^# `          【参考文献】6 G1 v" ^1 z& T8 Y* U' k: a/ m! L
$ u+ _2 x" d+ {& C
" f4 o' y/ ^- w5 j3 I' w% o
Smith AG, Heath JK, Donaldson DD et al. Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature 1988;336:688¨C690.
# {2 o" Y  J7 p, @  D9 {* @
& j+ K$ I* L# K6 o: r9 x4 wWilliams RL, Hilton DJ, Pease S et al. Myeloid leukaemia inhibitory factor maintains the developmental potential of embryonic stem cells. Nature 1988;336:684¨C687.
/ f7 n% N4 D3 \" ~6 {2 x
+ b3 b! U/ C8 u* E/ U. }" U+ [( eHeinrich PC, Behrmann I, Haan S et al. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J 2003;374:1¨C20.6 M8 v3 |8 w2 Z$ ~/ J! J. h
* W% q: c% C" r7 e! O
Stahl N, Farruggella TJ, Boulton TG et al. Choice of STATs and other substrates specified by modular tyrosine-based motifs in cytokine receptors. Science 1995;267:1349¨C1353.
) ~0 ]$ Y4 t2 h2 n5 J5 O
6 T, w' Y( j. n7 b0 ?! `5 v. eRaz R, Lee CK, Cannizzaro LA et al. Essential role of STAT3 for embryonic stem cell pluripotency. Proc Natl Acad Sci U S A 1999;96: 2846¨C2851.
- q) t# a( V. |) u; ]3 F  z7 x
% Z, I- ]' T9 a' IMatsuda T, Nakamura T, Nakao K et al. STAT3 activation is sufficient to maintain an undifferentiated state of mouse embryonic stem cells. EMBO J 1999;18:4261¨C4269.+ L7 b& }5 K1 I  R( ^* ?  \3 F

- g# a; N8 U5 {: P" h7 t/ u- UNiwa H, Burdon T, Chambers I et al. Self-renewal of pluripotent embryonic stem cells is mediated via activation of STAT3. Genes Dev 1998;12:2048¨C2060.
+ e' Q* C9 R$ C9 h& x. h3 x# Y3 W6 g( ?- @: N, A
Schaper F, Gendo C, Eck M et al. Activation of the protein tyrosine phosphatase SHP2 via the interleukin-6 signal transducing receptor protein gp130 requires tyrosine kinase Jak1 and limits acute-phase protein expression. Biochem J 1998;335:557¨C565.. n& d) H- k, _! N8 ~6 s4 s% l

# E! o% V: q2 b7 rFukada T, Hibi M, Yamanaka Y et al. Two signals are necessary for cell proliferation induced by a cytokine receptor gp130: Involvement of STAT3 in anti-apoptosis. Immunity 1996;5:449¨C460.
# d& K5 S! g+ [# s8 F9 s) ~/ c- q- e
Burdon T, Stracey C, Chambers I et al. Suppression of SHP-2 and ERK signalling promotes self-renewal of mouse embryonic stem cells. Dev Biol 1999;210:30¨C43.; Q8 T( D& ]) B% L! I3 B( s0 p

; Q, f6 w% Z$ N$ eQu CK, Feng GS. Shp-2 has a positive regulatory role in ES cell differentiation and proliferation. Oncogene 1998;17:433¨C439.
6 O" f. f* q) ^* R, {1 T
1 q" T8 r" W  k6 ?Nicholson SE, De Souza D, Fabri LJ et al. Suppressor of cytokine signaling-3 preferentially binds to the SHP-2-binding site on the shared cytokine receptor subunit gp130. Proc Natl Acad Sci U S A 2000;97:6493¨C6498.( N$ }! ~6 n+ l

. X# w! G, ], M  z/ c7 BKrebs DL, Hilton DJ. SOCS proteins: Negative regulators of cytokine signaling. STEM CELLS 2001;19:378¨C387.$ l& ~# {: G  e4 P+ s

. h; b5 u/ t3 pSchmitz J, Weissenbach M, Haan S et al. SOCS3 exerts its inhibitory function on interleukin-6 signal transduction through the SHP2 recruitment site of gp130. J Biol Chem 2000;275:12848¨C12856.
! [9 n. F: ~8 X) v- C" Z! x% ?: z  I) \! j- @% [' ^8 d. A1 ?
Kamura T, Sato S, Haque D et al. The Elongin BC complex interacts with the conserved SOCS-box motif present in members of the SOCS, Ras, WD-40 repeat, and ankyrin repeat families. Genes Dev 1998;12:3872¨C3881.
! A# V3 I0 U1 g4 `; I& c4 ?, \
$ P" y. D; \2 @Zhang JG, Farley A, Nicholson SE et al. The conserved SOCS box motif in suppressors of cytokine signalling binds to elongins B and C and may couple bound proteins to proteasomal degradation. Proc Natl Acad Sci U S A 1999;96:2071¨C2076.
0 w' n+ ~* m" B5 s+ A7 A5 g7 E0 Z. {3 N8 w! `, v# [! h
Kamura T, Maenaka K, Kotoshiba S et al. VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases. Genes Dev 2004;18:3055¨C3065.
% R/ \9 n1 m0 G. t$ H) c" ^2 _5 U
Rui L, Yuan M, Frantz D et al. SOCS-1 and SOCS-3 block insulin signalling by ubiquitin-mediated degradation of IRS1 and IRS2. J Biol Chem 2002;277:42394¨C42398.* F  h! T7 R9 _4 G# ^
. @$ G! Z1 e9 s  j3 \% v
Marine JC, McKay C, Wang D et al. SOCS3 is essential in the regulation of fetal liver erythropoiesis. Cell 1999;98:617¨C627., a- b' \. _8 u% k, F

# C( a! ^  n5 O# d  B5 J, K% H  E5 WRoberts AW, Robb L, Rakar S et al. Placental defects and embryonic lethality in mice lacking suppressor of cytokine signalling 3. Proc Natl Acad Sci U S A 2001;98:9324¨C9329.
% ^/ `, C6 j( m1 `* }) }/ ~( k6 J4 J) n5 X7 Y# A
Takahashi Y, Carpino N, Cross JC et al. SOCS3: An essential regulator of LIF receptor signalling in trophoblast giant cell differentiation. EMBO J 2003;22:372¨C384.
' j6 u# G0 q  f* X
. R0 W$ i; z+ ACroker BA, Krebs DL, Zhang JG et al. SOCS3 negatively regulates IL-6 signalling in vivo. Nat Immunol 2003;4:540¨C545.$ T) s: Q, b+ T

( R4 a( h* {0 G4 PLang R, Pauleau AL, Parganas E et al. SOCS3 regulates the plasticity of gp130 signaling. Nat Immunol 2003;4:546¨C550.5 R3 l, ?" p( E% j9 ~
- {7 G* [- y* W: O
Yasukawa H, Ohishi M, Mori H et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nat Immunol 2003; 4:551¨C556.
- h) S, }& c& N. ?
. D  y6 ^+ l8 bDuval D, Reinhardt B, Kedinger C et al. Role of suppressors of cytokine signalling (SOCS) in leukemia inhibitory factor (LIF)-dependent embryonic stem cell survival. FASEB J 2000;14:1577¨C1584.
2 L; i, e, l9 @' k
3 Z3 r: D6 _2 nChambers I, Smith A. Self-renewal of teratocarcinoma and embryonic stem cells. Oncogene 2004;23:7150¨C7160.( D8 s, A0 w' M+ k9 j. x% C

+ ^  j3 [2 L/ cLi Y, McClintick J, Zhong L et al. Murine embryonic stem cell differentiation is promoted by SOCS-3 and inhibited by the zinc finger transcription factor Klf4. Blood 2004;105:635¨C637.
; ]9 ~3 i# H8 {- m+ _9 t% d% l! U4 u
Voss AK, Thomas T, Gruss P. Germ line chimeras from female ES cells. Exp Cell Res 1997;230:45¨C49.
  }2 K" V) p4 A/ r6 T* m% i; ^9 ~$ l/ B% l' H" H: z4 s
Barnett LD, Kontgen F. Gene targeting in a centralized facility. Methods Mol Biol 2001;158:65¨C82.
7 M4 I) c4 g, K* j8 E3 }1 N
; k4 f1 n, Y5 |; q9 L  RKennedy M, Keller GM. Hematopoietic commitment of ES cells in culture. Methods Enzymol 2003;365:39¨C59.
6 q5 I' S. @4 }' k" ~. ?$ a' n, p* L4 A, S4 O
Elefanty AG, Robb L, Birner R et al. Hematopoietic-specific genes are not induced during in vitro differentiation of scl-null embryonic stem cells. Blood 1997;90:1435¨C1447.# T; q0 t( ?0 |, z# Q9 H* {2 D

8 K5 t. A- U. `3 f6 B( KHart AH, Hartley L, Ibrahim M et al. Identification, cloning and expression analysis of the pluripotency promoting Nanog genes in mouse and human. Dev Dyn 2004;230:187¨C198.
) a& P- @% K+ p! D) }: V# c7 A7 ?0 ^6 |) Q7 k* p' o2 e3 u
Robb L, Hartley L, Begley CG et al. Cloning, expression analysis, and chromosomal localization of murine and human homologues of a Xenopus mix gene. Dev Dyn 2000;219:497¨C504.8 S! L( [/ {( K. s

' _# h( W/ k5 X% h$ HMaye P, Becker S, Siemen H et al. Hedgehog signalling is required for the differentiation of ES cells into neurectoderm. Dev Biol 2004;265:276¨C290.
4 R, j( |  l8 ~! m# v/ z5 u
" f3 F3 ?% z% ~Keller G, Kennedy M, Papayannopoulou T et al. Hematopoietic commitment during embryonic stem cell differentiation in culture. Mol Cell Biol 1993;13:473¨C486.
; w2 h& ]1 m+ x! @) G: P1 q; s) I9 u% m) _0 |8 Q* q4 d. N
Fujikura J, Yamato E, Yonemura S et al. Differentiation of embryonic stem cells is induced by GATA factors. Genes Dev 2002;16:784¨C789.& `1 [3 O) ~$ D+ n; N
. B8 k0 t; \4 _  B, f+ x9 z: H
Ginis I, Luo Y, Miura T et al. Differences between human and mouse embryonic stem cells. Dev Biol 2004;269:360¨C380." h" b* \- K! c+ e3 k0 x+ g3 x9 o" Z
: X& @: S' u' z  \4 z& W0 {
Irizarry RA, Bolstad BM, Collin F et al. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 2003;31:e15.5 s( J/ p: H) t( T

& b$ r, d; B5 A9 B4 m* O1 v& J4 SSmyth GK. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 2004;3:1¨C26.7 X; ?& O3 P" ?6 n4 q  i# p
8 g/ j5 Z1 I' d$ D& {, j1 x1 `
Varlet I, Collignon J, Robertson EJ. Nodal expression in the primitive endoderm is required for specification of the anterior axis during mouse gastrulation. Development 1997;124:1033¨C1044.7 n% Q5 Z+ Y+ B! F4 x; i

2 T( s2 h: s# }1 zHart AH, Hartley L, Sourris K et al. Mixl1 is required for axial mesendoderm morphogenesis and patterning in the murine embryo. Development 2002;129:3597¨C3608.
) l5 e7 S7 c- ]2 ^
* a/ G0 I' r, v# uRobb L, Elwood NJ, Elefanty AG et al. The scl gene product is required for the generation of all hematopoietic lineages in the adult mouse. EMBO J 1996;15:4123¨C4129./ q( f+ u# B( m/ R

$ ?  m' t( h1 l0 [5 b, T2 m  WSzabo P, Mann JR. Expression and methylation of imprinted genes during in vitro differentiation of mouse parthenogenetic and androgenetic embryonic stem cell lines. Development 1994;120:1651¨C1660.5 [) p+ \) M# @( k2 S7 e/ k3 d

3 J+ |1 s# t  b* \% rJetten AM, Jetten ME, Sherman MI. Analyses of cell surface and secreted proteins of primary cultures of mouse extraembryonic membranes. Dev Biol 1979;70:89¨C104.
' z8 F4 C7 F3 V1 w3 G5 I" Z
" W5 s8 F. x& A5 A2 u- DTam PP, Kanai-Azuma M, Kanai Y. Early endoderm development in vertebrates: Lineage differentiation and morphogenetic function. Curr Opin Genet Dev 2003;13:393¨C400.
! |' W, Y( q  I, Q- D  J* q  s$ F6 z1 W) d) w
Kubo A, Shinozaki K, Shannon JM et al. Development of definitive endoderm from embryonic stem cells in culture. Development 2004;131: 1651¨C1662.0 o6 x( X+ k' G- S6 D" T
& `$ p6 R4 M0 K* G: x) Y3 e% L3 t- e( y
Smedberg JL, Smith ER, Capo-Chichi CD et al. Ras/MAPK pathway confers basement membrane dependence upon endoderm differentiation of embryonic carcinoma cells. J Biol Chem 2002;277:40911¨C40918.% Y# |5 Z2 Q) {1 X0 \

- T7 r% o% r' u, h4 T1 |Futaki S, Hayashi Y, Yamashita M et al. Molecular basis of constitutive production of basement membrane components. Gene expression profiles of Engelbreth-Holm-Swarm tumor and F9 embryonal carcinoma cells. J Biol Chem 2003;278:50691¨C50701.
) l% L; U0 {! |* x
0 B" z9 `* p% p/ kStevens LC. The biology of teratomas. Adv Morphog 1967;6:1¨C31./ a  z, s2 |; f% k. ]

. V8 s; l% i0 W9 }- TAlessi DR, Cuenda A, Cohen P et al. PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J Biol Chem 1995;270:27489¨C27494.- K4 q* R" F; `. Q
$ d, G& d  y% v% s( V
Favata MF, Horiuchi KY, Manos EJ et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem 1998; 273:18623¨C18632.
4 M2 w# g. ~: S2 U' ^+ ^7 T0 f2 }" H, N% q* r) f8 i
Cacalano NA, Sanden D, Johnston JA. Tyrosine-phosphorylated SOCS-3 inhibits STAT activation but binds to p120 RasGAP and activates Ras. Nat Cell Biol 2001;3:460¨C465.; ^' u5 F9 k) D

5 ~: V5 R5 O  d3 gOhtani T, Ishihara K, Atsumi T et al. Dissection of signalling cascades through gp130 in vivo: Reciprocal roles for STAT3- and SHP2-mediated signals in immune responses. Immunity 2000;12:95¨C105., J  p. q5 ?5 b8 d0 n2 w
0 k0 N( I% ?; Y8 S0 w
Atsumi T, Ishihara K, Kamimura D et al. A point mutation of Tyr-759 in interleukin 6 family cytokine receptor subunit gp130 causes autoimmune arthritis. J Exp Med 2002;196:979¨C990., s+ F1 s" P) I! Y* D, Z
1 j! b/ d  h+ Y5 ~
Tebbutt NC, Giraud AS, Inglese M et al. Reciprocal regulation of gastrointestinal homeostasis by SHP2 and STAT-mediated trefoil gene activation in gp130 mutant mice. Nat Med 2002;8:1089¨C1097.! B/ e$ A& K7 A  o2 y3 @

4 U. p: o0 W6 h  ?. O) v3 O4 _Ernst M, Inglese M, Waring P et al. Defective gp130-mediated signal transducer and activator of transcription (STAT) signalling results in degenerative joint disease, gastrointestinal ulceration, and failure of uterine implantation. J Exp Med 2001;194:189¨C203.
& \. ]/ q/ {! j0 U" S1 u
6 ~- S. R& {6 |" U( aSmith AG. Embryo-derived stem cells: Of mice and men. Annu Rev Cell Dev Biol 2001;17:435¨C462.
' S6 U1 i* T$ i5 k% F7 ~
+ G) C4 O% X7 L; YBurdon T, Smith A, Savatier P. Signalling, cell cycle and pluripotency in embryonic stem cells. Trends Cell Biol 2002;12:432¨C438.4 Y9 F2 t0 p2 h+ r( M
" y8 u1 [: |2 o
Cheng AM, Saxton TM, Sakai R et al. Mammalian Grb2 regulates multiple steps in embryonic development and malignant transformation. Cell 1998;95:793¨C803.% W/ L8 Q4 m/ C1 g4 g9 F# p
; b' P  o7 s4 ^1 ?8 E
Yoshida-Koide U, Matsuda T, Saikawa K et al. Involvement of Ras in extraembryonic endoderm differentiation of embryonic stem cells. Biochem Biophys Res Commun 2004;313:475¨C481.: x$ b6 ~/ i. b; }+ }  x0 @
9 l2 S+ v, U. P& n# f% J
Viswanathan S, Benatar T, Mileikovsky M et al. Supplementation-dependent differences in the rates of embryonic stem cell self-renewal, differentiation, and apoptosis. Biotechnol Bioeng 2003;84:505¨C517.
% X) d, W: a4 p4 g( K, f* |
) n8 o2 s+ y3 `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.
$ g, |# _+ t6 S3 J% N) n/ j
6 ]; y# j1 U( O( I+ V9 S# y2 d$ |Sato N, Meijer L, Skaltsounis L et al. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signalling by a pharmacological GSK-3-specific inhibitor. Nat Med 2004;10:55¨C63.
: X9 F3 H4 I2 I  i0 _
% ^; d$ X: F; s. F# r) IChambers 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 |/ ~1 a" q7 I( w1 K* h- B" P
" Q" p0 u2 L4 \6 P% Z
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.. g  G2 |7 d* q, i7 [

  Q1 ?5 c4 Q4 ?: ]0 n3 `Stewart CL, Kaspar P, Brunet LJ et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature 1992;359:76¨C79.0 H  e! Z. y+ A  U% s- D1 {5 _

/ C" ?+ j& T3 j' nLi M, Sendtner M, Smith A. Essential function of LIF receptor in motor neurons. Nature 1995;378:724¨C727.
2 C0 ?& d5 `* E# g8 w( O6 f* `6 k/ Y4 z7 t8 g7 j$ u
Ware CB, Horowitz MC, Renshaw BR et al. Targeted disruption of the low-affinity leukemia inhibitory factor receptor gene causes placental, skeletal, neural and metabolic defects and results in perinatal death. Development 1995;121:1283¨C1299.
% H9 F8 a5 b* M. A) p
# v# k& ~/ V9 V0 s4 [( cYoshida K, Taga T, Saito M et al. Targeted disruption of gp130, a common signal transducer for the interleukin 6 family of cytokines, leads to myocardial and hematological disorders. Proc Natl Acad Sci U S A 1996;93:407¨C411.. o) g: X8 h# \' w5 q+ y8 x% {

% t3 K; [4 v" }$ Z4 X, `6 hNichols J, Chambers I, Taga T et al. Physiological rationale for responsiveness of mouse embryonic stem cells to gp130 cytokines. Development 2001;128:2333¨C2339.

Rank: 2

积分
166 
威望
166  
包包
1997  
沙发
发表于 2015-6-24 15:11 |只看该作者
干细胞研究还要面向临床

Rank: 2

积分
77 
威望
77  
包包
1730  
藤椅
发表于 2015-7-12 18:42 |只看该作者
不错的东西  持续关注  

Rank: 2

积分
80 
威望
80  
包包
1719  
板凳
发表于 2015-8-7 21:33 |只看该作者
干细胞之家微信公众号
这个贴不错!!!!!看了之后就要回复贴子,呵呵  

Rank: 2

积分
76 
威望
76  
包包
1772  
报纸
发表于 2015-8-28 19:42 |只看该作者
谢谢分享  

Rank: 2

积分
77 
威望
77  
包包
1964  
地板
发表于 2015-9-2 15:18 |只看该作者
不错不错,我喜欢看  

Rank: 2

积分
80 
威望
80  
包包
1719  
7
发表于 2015-9-13 19:03 |只看该作者
帮你项项吧  

Rank: 2

积分
79 
威望
79  
包包
1769  
8
发表于 2015-10-10 18:51 |只看该作者
朕要休息了..............  

Rank: 2

积分
98 
威望
98  
包包
1756  
9
发表于 2015-11-19 20:26 |只看该作者
干细胞库  

Rank: 2

积分
79 
威望
79  
包包
1769  
10
发表于 2015-11-24 21:43 |只看该作者
不看白不看,看也不白看  
‹ 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, 2024-5-4 06:42

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.