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: 395846|回复: 251
go

Glycogen synthase kinase-3 is an endogenous inhibitor of Snail transcr [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
3465  
楼主
发表于 2009-3-6 08:41 |只看该作者 |倒序浏览 |打印
1 Department of Pathology, Division of Cancer Biology and Angiogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
& p9 S& A! T! d
/ h3 C: I1 ?' U6 Y& I/ I2 Unitat de Biologia Cellular i Molecular, Institut Municipal d'Investigaci車 Mèdica, Universitat Pompeu Fabra, 08003 Barcelona, Spain
) w% |% o3 O; H6 L7 k# Y7 W3 e& R; A" n
Correspondence to Robin E. Bachelder: rbacheld@bidmc.harvard.edu
+ w6 ^* ~; C' S" |: c
5 d8 t8 T0 I; G2 m5 u  l. oAbstract! `" B) ]! d3 `6 a% f# S

3 z" q' j6 {& D+ s5 aWe report that the activity of glycogen synthase kinase-3 (GSK-3) is necessary for the maintenance of the epithelial architecture. Pharmacological inhibition of its activity or reducing its expression using small interfering RNAs in normal breast and skin epithelial cells results in a reduction of E-cadherin expression and a more mesenchymal morphology, both of which are features associated with an epithelial–mesenchymal transition (EMT). Importantly, GSK-3 inhibition also stimulates the transcription of Snail, a repressor of E-cadherin and an inducer of the EMT. We identify NFB as a transcription factor inhibited by GSK-3 in epithelial cells that is relevant for Snail expression. These findings indicate that epithelial cells must sustain activation of a specific kinase to impede a mesenchymal transition.
7 i+ S% t6 X& w8 n1 v& D
* }( W6 C2 ?2 V& F$ Z9 qAbbreviations used in this paper: EMT, epithelial–mesenchymal transition; GSK-3, glycogen synthase kinase-3; siRNA, small interfering RNA.2 g  H, N6 L; u( j) M+ Y
' k6 z/ @" N4 ]5 r( {
Introduction
6 Y7 m7 D  P" d' q6 ~0 ?. y" I
. _% |1 N* z' X7 B/ EThe distinct architecture of epithelia is derived from the interactions of epithelial cells with each other, with the underlying basement membrane, and, less directly, with mesenchymal cells in the stroma. Within this complex set of interactions, a prime determinant of epithelial structure is E-cadherin, a transmembrane protein that mediates Ca  -dependent, homophilic intercellular adhesion (Thiery, 2002; Nelson and Nusse, 2004). The central role of E-cadherin in epithelia is evidenced by the fact that loss of either its expression or function results in the dissolution of the epithelial architecture and the acquisition of a mesenchymal phenotype. This process, referred to as the epithelial–mesenchymal transition (EMT), occurs in the contexts of development and tumor progression (Thiery, 2002).
9 S) s3 V  Y: t+ i% p8 |; a6 ?& P* `
The central importance of E-cadherin for epithelial architecture leads to the novel hypothesis that epithelial cells may support signaling pathways that preserve E-cadherin expression as a means of preventing an EMT. In this connection, we were intrigued by reports that glycogen synthase kinase-3 (GSK-3), a ubiquitously expressed protein serine kinase, is active in resting epithelial cells (Papkoff and Aikawa, 1998; Murray et al., 1999), but that its function in epithelial biology had not been defined. Our assessment of GSK-3 function in epithelial cells revealed that its activity is essential for maintaining epithelial structure because it maintains the expression of E-cadherin. Inhibition of GSK-3 activity or expression results in a bona fide EMT. Moreover, we report that one mechanism by which GSK-3 maintains E-cadherin expression is by inhibiting the transcription of Snail, a zinc finger transcriptional repressor of E-cadherin that is absent in epithelial cells but expressed in tumors (Batlle et al., 2000; Cano et al., 2000; Blanco et al., 2002).
9 Z- v% j7 u, H& g" Z7 P$ Q5 D) y8 s+ [$ G# }: G
Results and discussion
3 y0 M8 Y0 B# L" w0 `% t
$ J+ t; w8 S2 D2 L  V' t( c4 yInitially, we assessed the effects of inhibiting GSK-3 activity in normal breast epithelial cells (MCF10A) using SB415286, a highly specific, small molecule inhibitor of GSK-3 (Coghlan et al., 2000). The ability of SB415286 to inhibit GSK-3 activity was evidenced by the increased levels of cyclin D1, a protein subject to GSK-3–dependent proteolysis (Diehl et al., 1998), in SB415286-treated MCF10A cells, relative to those treated with DMSO (Fig. 1 A). Inhibiting GSK-3 activity also disrupted the epithelial morphology of these cells, as evidenced by the loss of cell–cell contacts (Fig. 1 A).
) u6 [- b( v- x7 x
/ G+ \; a! n$ T5 WFigure 1. GSK-3 maintains the epithelial phenotype. (A) MCF10A cells were incubated with either DMSO or 25 μM SB415286 (Biosource International) in 0.5% FBS-containing medium. Cell morphology was assessed after 72 h by phase contrast microscopy. Expression of Cyclin D1, Stat-1, E-cadherin, and vimentin was assessed by immunoblotting. (B) HaCaT cells were incubated with DMSO or SB415286, and E-cadherin, Stat-1, and cyclin D1 expression was assessed as indicated in A. Similar results were obtained in four independent experiments. (C) E-cadherin and GAPDH mRNA levels were determined after 48 h of drug treatment by RT-PCR. (D) MCF10A cells were cotransfected with GSK-3– and GSK-3?–specific inhibitory RNA pools ( ? siRNA) or a nonspecific control pool (Scr). RNA was extracted from these cells 48 h after transfection, and E-cadherin and GAPDH mRNA levels were determined by RT-PCR. GSK-3 and Stat-1 expression was assessed by immunoblotting.  b( e0 M! K$ s0 W" v' E, |& {
4 r3 |7 R1 |- |2 H! ^* l! J3 G
Loss of E-cadherin and expression of mesenchymal proteins are defining steps in the EMT. Based on our observation that inhibition of GSK-3 activity reduced cell–cell contacts in MCF10A epithelial cells, we hypothesized that GSK-3 may be a regulator of E-cadherin expression and an inhibitor of the EMT. Supporting these hypotheses, the treatment of MCF10A cells with SB415286 reduced the expression of total cellular E-cadherin protein significantly and induced the expression of the mesenchymal protein vimentin (Fig. 1 A) without influencing cell viability (not depicted). Together, these data indicate that epithelial cells in which GSK-3 activity has been inhibited manifest changes characteristic of an EMT.
- G, {* U1 n0 `& O4 b$ j! m7 S( Z
The effects of GSK-3 inhibition can be generalized to other epithelial cells, as demonstrated by the significantly reduced levels of E-cadherin protein in HaCaT skin cells that had been incubated with the GSK-3 inhibitor (Fig. 1 B). This treatment also increased cyclin D1 expression, indicating the efficacy of SB415286 in HaCaT cells (Fig. 1 B). GSK-3 inhibition also reduced the levels of E-cadherin mRNA in GSK-3 inhibitor–treated HaCaT and MCF10A cells, relative to controls (Fig. 1 C). We implemented a small interfering RNA (siRNA) strategy to reduce expression of both the  and ? isoforms of GSK-3 in MCF10A cells. Cells were transfected with a pool of eight siRNAs, targeting unique regions of GSK-3 and GSK-3? genes, or with a pool of nonspecific sequences (Fig. 1 D, Scr). Relative to the control pool, the GSK-3 and GSK-3? siRNA pool significantly decreased expression of GSK-3 and GSK-3? proteins (Fig. 1 D). Similar to the effect of the GSK-3 inhibitor, GSK-3–specific siRNAs markedly reduced E-cadherin mRNA (Fig. 1 D).
( a1 C6 E5 {& d2 ?4 L  c; ~4 s* F2 X- z
We next sought to determine the mechanism by which GSK-3 regulates E-cadherin expression. Snail, a member of the zinc finger family of transcriptional repressors, is an established suppressor of E-cadherin transcription, and its activity is an important determinant of the EMT, in the contexts of both mesoderm development (Carver et al., 2001) and tumor progression (Batlle et al., 2000; Cano et al., 2000). Snail is absent in epithelial cells but expressed in tumors, and its expression has been shown to correlate inversely with tumor grade (Blanco et al., 2002). We investigated whether GSK-3 regulates E-cadherin expression by repressing Snail expression. Inhibition of GSK-3 activity in either MCF10A or HaCaT cells significantly increased Snail mRNA levels (Fig. 2 A). Snail mRNA levels were also elevated in epithelial cells transfected with GSK-3–specific siRNAs (Fig. 2 A). As evidence that GSK-3 inhibition alters Snail transcription, we observed increased activity of a Snail promoter–driven reporter gene in HaCaT cells treated with SB415286, relative to that measured in DMSO-treated cells (Fig. 2 B). Increased levels of Snail protein were also detected in cells treated with the GSK-3 inhibitor (Fig. 2 C).+ s- W  m* a2 I3 D

/ R, ?- ]2 X. n- R3 ]1 x; {Figure 2. GSK-3 inhibits Snail transcription in epithelial cells. (A) MCF10A and HaCaT cells were either incubated with 25 μM of the GSK-3 inhibitor SB415286 or with DMSO in 0.5% FBS-containing medium. Alternatively, these cells were transfected with GSK-3– and GSK-3?–specific inhibitory RNA pools ( ? siRNA) or a nonspecific control pool (Scr). Snail and GAPDH mRNA levels were assessed by reverse transcription, followed by PCR. GSK-3 and Stat-1 expression was assessed by immunoblotting. (B) HaCaT cells were cotransfected with a Snail promoter–driven and a control ?-galactosidase reporter construct. These transfectants were then incubated with DMSO or SB415286, as described in A, for 24 h. Data are reported as relative luciferase activity (normalized to ?-galactosidase activity), ± SD, and are representative of three independent experiments. (C) MCF10A and HeLa cells were treated with SB415286 as described in A, and Snail expression was assessed by immunofluorescence.
3 K# X- V: F9 _6 p$ \/ w
6 Z+ Y/ w2 @1 J: [Because of our recent finding that NFB drives Snail expression (Barbera et al., 2004), we explored whether GSK-3 inhibits Snail transcription by repressing NFB activity. Indeed, inhibition of GSK-3 activity in HaCaT cells stimulated NFB-dependent reporter gene expression (Fig. 3 A). In addition, we observed significantly decreased levels of IB, an inhibitor of NFB, in SB415286-treated MCF10A cells (Fig. 3 B). Finally, an NFB inhibitor (SN50) suppressed the ability of SB415286 to induce Snail expression (Fig. 3 C). Together, these data indicate that the NFB pathway is inhibited by GSK-3 in epithelial cells, which results in the silencing of Snail expression.
/ H, c# ^1 ]9 G  g3 H- G; J: {; d: s4 R6 I' J  q0 K
Figure 3. GSK-3 inhibits NFB, an activator of Snail transcription. (A) HaCaT cells were transfected transiently with a luciferase construct driven by NFB binding sites in addition to a control renilla luciferase reporter gene. These transfectants were then incubated with DMSO or SB415286, as described for Fig. 2, for 15 h. Data are reported as relative luciferase activity (normalized to renilla luciferase activity), ± SD, and are representative of two independent experiments. (B) MCF10A cells were incubated with DMSO or SB415286 as described for Fig. 2. Equivalent amounts of total cellular protein were extracted from these cells, subjected to SDS-PAGE, and immunoblotted with an IB- or actin-specific antibody. Results are representative of those obtained in two independent experiments. (C) MCF10A cells were incubated for 2 h with an NFB-specific inhibitor (SN50; Calbiochem) or control peptide (SN50M; Calbiochem ). These cells were then incubated with DMSO or 25 μM SB415286. RNA was isolated after 15 h, and the levels of Snail and GAPDH were determined by RT-PCR. Similar results were obtained in three independent experiments./ u3 O6 p4 C/ _7 q) i
/ a8 e1 }1 _- |9 G: M0 A4 J
The data presented here indicate that GSK-3, a kinase that is active in resting epithelial cells (Papkoff and Aikawa, 1998; Murray et al., 1999), is a critical determinant of epithelial structure and a suppressor of the EMT. This finding implies that epithelial cells must sustain activation of a specific kinase to prevent an EMT, a mechanism distinct from that suggested by previous studies, which have shown that activation of kinases such as Akt (Grille et al., 2003) and ILK (Oloumi et al., 2004) can promote an EMT. An important implication of our findings is that endogenous suppressors of GSK-3, such as Wnt and PI3-kinase, which are frequently activated in carcinoma cells (Woodgett, 2001), may also inhibit E-cadherin transcription and promote an EMT. Our observation that inhibition of both GSK-3 and GSK-3? isoforms increased Snail expression in epithelial cells (Fig. 2 A), whereas inhibition of either isoform alone had no effect on Snail levels (not depicted), indicates that these isoforms may serve redundant functions in maintaining epithelial architecture. Thus, we hypothesize that GSK-3 and GSK-3? isoforms serve redundant functions in epithelial cells, and that mice deficient for both GSK-3 and GSK-3? expression will exhibit prominent defects in epithelial structure and function.
" ~4 j/ V7 E1 W5 A' p2 y1 d5 i0 d8 y4 n
* W. K. D: S0 ]9 {The identification of NFB as a novel target of GSK-3 activity that is relevant to the EMT is of considerable interest in light of a recent report that established NFB as a central mediator of the EMT (Huber et al., 2004). However, this study did not define transcriptional targets of NFB that are important for the EMT. Clearly, Snail is a prime candidate for such an NFB target.: Q# t- [6 p! n% l
( j% i) N/ D8 J8 U4 t) W; n
Although we identify NFB and Snail as novel targets of GSK-3 activity that are relevant to the EMT, it is likely that multiple GSK-3 substrates participate in the regulation of epithelial structure. Upon its phosphorylation by GSK-3, ?-catenin, a protein that has been implicated in mesenchymal transitions (Kim et al., 2002; Liebner et al., 2004), is degraded by the proteosome pathway, resulting in its inability to stimulate TCF/LEF transcription factors (Beals et al., 1997). TCF/LEF can induce the expression of genes that influence the EMT, such as vimentin (Gilles et al., 2003), the levels of which are elevated in epithelial cells treated with the GSK-3 inhibitor (Fig. 1 A). Thus, multiple signaling pathways that control the EMT are likely to be regulated by GSK-3, substantiating our hypothesis that this kinase is a central regulator of epithelial structure and function.
( M9 R( f  D2 \1 k" y6 q+ C8 j' ?2 }, v3 L  f/ v4 g
It is important to mention that Snail was recently identified as a direct target of GSK-3 kinase activity (Zhou et al., 2004), and the two GSK-3 phosphorylation sites identified were shown to influence Snail protein stability and localization in tumor cells. In contrast, the present study indicates that, in epithelial cells, GSK-3 impedes Snail transcription. The data presented here provide one explanation for why epithelial cells, which are characterized by high basal GSK-3 activity (Papkoff and Aikawa, 1998; Murray et al., 1999), express negligible levels of Snail mRNA (Fig. 2; Dom赤nguez et al., 2003; Peinado et al., 2003). The combined abilities of GSK-3 to block Snail transcription, promote Snail degradation, and prevent Snail nuclear localization indicate that this kinase plays a central role in regulating Snail expression. Future studies addressing the importance of posttranslational control of Snail by GSK-3 in epithelial cells will be crucial for establishing the relevance of this mode of regulation for the EMT.
- B' u' V. t! S/ l, |1 b5 X" }4 R  t, r( h/ p3 @
GSK-3 is a central target for the development of therapeutics because it has been implicated in numerous pathologies, including diabetes and neurologic disorders (Woodgett, 2001). Our findings suggest that this kinase is active in epithelial cells for a very important reason, namely to prevent the acquisition of a mesenchymal phenotype. Because reduced E-cadherin expression has been reported to be characteristic of some malignant tumors (Bringuier et al., 1993; Hirohashi, 1998; Kowalski et al., 2003), our findings indicate that inhibition of GSK-3, an enzyme that maintains E-cadherin expression, will not prove to be a reasonable therapeutic strategy because it has the potential to alter epithelial function significantly.
  k! Y  X- @$ N6 S, P, S0 T9 Q7 L4 M- J' t* g, S8 J
Materials and methods
: S. \9 \3 `3 P3 U- q) g4 B* A
8 Y- A8 @9 z! H  G0 f* zCells
# ^# x* U: f' P8 m2 w7 g: T* F+ {2 L3 S8 D6 {3 n, ~: {
HaCat and HeLa cells were provided by A. Toker (Beth Israel Deaconess Medical Center, Boston, MA). MCF10A cells were provided by J. Brugge (Harvard Medical School, Boston, MA).
0 W' a# {8 i: A5 V( F9 e% o
5 K3 ^0 u& E* z  m! e! uAnalysis of protein expression
# B8 G' g. X- O2 J: I4 k% o2 q$ z8 I# G
Cells were extracted in RIPA buffer. Equivalent amounts of total cellular protein extracted from these cells were subjected to SDS-PAGE and transferred to nitrocellulose. The antibodies used for immunoblotting were as follows: GSK-3 monoclonal antibody (Upstate Biotechnology), rabbit anti–cyclin D1 (Santa Cruz Biotechnology, Inc.), rabbit anti–Stat-1 (Santa Cruz Biotechnology, Inc.), mouse anti–E-cadherin (Santa Cruz Biotechnology, Inc.), rabbit antivimentin (Santa Cruz Biotechnology, Inc.), mouse anti-–IB  (Cell Signaling), and HRP-conjugated secondary antibodies (Pierce Chemical Co.).
1 l2 z" m% y# f% M; d
/ e. ^; I2 n; M9 f' B3 L  C5 T. q7 rLuciferase assays
2 P9 r! M5 I- `& X! @4 R  _
5 c+ d* l7 B# H; @0 |HaCaT cells were cotransfected transiently using Lipofectin (Life Technologies) with a Snail promoter luciferase reporter construct (composed of human snail promoter sequences –869 to 59) and a control ?-galactosidase reporter construct (pCS2-(n)-?gal; Promega). For NFB studies, HaCaT cells were transfected with a luciferase construct controlled by sequential NFB binding sites (pNF-B-Luc; CLONTECH Laboratories, Inc.) and a control renilla luciferase construct (pRL-TK renilla luciferase; Promega). After incubation for 12 h, cells were treated with the indicated drugs (Fig. 3 A) for an additional 15 h. Luciferase and ?-galactosidase activities were measured according to the manufacturer's instructions (Promega) using a luminometer and a UV/Vis spectrophotometer, respectively. Data are reported (Fig. 3 A) as the mean (± SD) relative promoter activity obtained from triplicate wells.2 G# C+ N6 a/ j) K' k% X( J
' u2 c6 I. z- a( x2 U! F- ~
Analysis of mRNA expression8 e% L  c- l& d

  H8 Y9 o* u2 P3 N6 Q  ]" x6 a$ umRNA was purified from the indicated cell lines using the RNEasy kit (QIAGEN) according to the manufacturer's recommended protocol. 2 μg RNA was added to RT-PCR reactions containing the indicated primers at a concentration of 0.6 μM. After a 55°C/30-min reverse transcription step, Snail mRNA was PCR amplified in 32 cycles for 1 min at each of the following temperatures: 94~, 62~, and 72°C. Likewise, E-cadherin mRNA was PCR amplified in 25 cycles for 1 min at each of the following temperatures: 94~, 55~, and 72°C. PCR products were analyzed on 1% agarose gels. The sequences of amplification primers were as follows: Snail forward, GGGCAGGTATGGAGAGGAAGA; Snail reverse, TTCTTCTGCGCTACTGCTGCG; E-cadherin forward, CAGCACGTACACAGCCCTAA; E-cadherin reverse, GCTGGCTCAAGTCAAAGTCC; GAPDH forward, CCTGGCCAAGGTCATCCATGAC; and GAPDH reverse, CATGTAGGCCATGAGGTCCACCAC.
: q) _& _  }. T7 a. j
, U6 G$ S" a! c  u& |. z: t8 X# SsiRNA experiments
1 ^4 i6 y6 W+ Y+ {5 C0 t$ K0 t9 D! f" `8 O$ b
GSK-3, GSK-3?, and control siRNA pools were designed and synthesized by Upstate Biotechnology. Cells at 60% confluence were transfected in penicillin/streptomycin-free medium with 20 μM of the indicated siRNA using TKO lipid (Mirus) according to the manufacturer's recommended protocol. Cells were harvested 48 h after transfection.
, o! E- S* y# Y' E4 t4 h  ]+ x9 }- M- d7 T: _/ X# _
Immunofluorescence
/ L1 `; f4 }% A. {
+ e0 r2 l( U' @% c+ G; tCells were fixed in 2% PFA, permeabilized in 0.2% Triton X-100 and 1 mM EGTA, blocked for 30 min in 1% albumin/5% donkey serum, and incubated overnight at 4°C with a Snail-specific (Dom赤nguez et al., 2003) or control rabbit serum in blocking buffer (at a 1:20 dilution). The cells were then incubated with a fluorescein-conjugated donkey anti–rabbit IgG (Jackson ImmunoResearch Laboratories) in blocking buffer (at a 1:150 dilution) for 30 min. Snail expression was analyzed using an inverted fluorescent microscope (Diaphot 300; Nikon).
( r7 d1 n; X1 s9 }* x0 {5 k* t, N4 L! V& Q; E/ @: L+ l; R9 X
Acknowledgments/ s7 s) Y; j7 }6 y7 a

' }( ]. m$ @: I; s5 b: WWe thank Drs. James Woodgett and Alex Toker for valuable discussion, and Xuena Lin for technical assistance.
* x+ c* d0 {  a& o& R/ k% K
4 o. @9 @9 k7 }2 N: NThis work was supported by National Institutes of Health grants CA89209 and CA107548 (to A.M. Mercurio) and CA93855 (to R.E. Bachelder).
: d5 S4 W' o( T2 w2 F7 g& ]3 o" ^0 H( k; |1 f) B" Y; {& _
Submitted: 10 September 2004
, T0 E3 G7 w+ ]0 B- A/ h) J% d( ~/ D2 H
Accepted: 5 November 2004! F; f: T& X/ ~$ a1 g2 J4 l

0 f7 _1 Y' a7 C8 l3 ~7 JReferences2 i8 C% y9 K0 v+ C
( t$ o( J# a/ |! H7 E) Z
Barbera, M.J., I. Puig, D. Dominguez, S. Julien-Grille, S. Guaita-Esteruelas, S. Peiro, J. Baulida, C. Franci, S. Dedhar, L. Larue, and A. Garcia De Herreros. 2004. Regulation of Snail transcription during epithelial to mesenchymal transition of tumor cells. Oncogene. 23:7345–7354.: n' P3 y4 O/ W5 v) B
2 T/ m3 P; q9 b. {$ _/ N4 i
Batlle, E., E. Sancho, C. Franci, D. Dominguez, M. Monfar, J. Baulida, and A. Garcia De Herreros. 2000. The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat. Cell Biol. 2:84–89.
$ }1 u( w& _7 @3 {3 j/ x% N9 E
9 r& k+ c. P) _9 lBeals, C.R., C.M. Sheridan, C.W. Turck, P. Gardner, and G.R. Crabtree. 1997. Nuclear export of NF-ATc enhanced by glycogen synthase kinase-3. Science. 275:1930–1934.
& w  q% F4 |+ H  b+ e& r
7 ]7 o/ f3 K9 e  NBlanco, M.J., G. Moreno-Bueno, D. Sarrio, A. Locascio, A. Cano, J. Palacios, and M.A. Nieto. 2002. Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene. 21:3241–3246.
1 T. i9 ?( I% c3 _& O' K: x; x; o# {: M" I
Bringuier, P.P., R. Umbas, H.E. Schaafsma, H.F. Karthaus, F.M. Debruyne, and J.A. Schalken. 1993. Decreased E-cadherin immunoreactivity correlates with poor survival in patients with bladder tumors. Cancer Res. 53:3241–3245.
6 b, [9 M9 T! _& A: p5 L
8 q9 F# z  n2 Y2 ?5 Z- c5 jCano, A., M.A. Perez-Moreno, I. Rodrigo, A. Locascio, M.J. Blanco, M.G. del Barrio, F. Portillo, and M.A. Nieto. 2000. The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat. Cell Biol. 2:76–83.  O; g. M* T" \2 v2 c3 {, A. Z
# Y* Q  w5 a; D1 Z+ ~( C9 w
Carver, E.A., R. Jiang, Y. Lan, K.F. Oram, and T. Gridley. 2001. The mouse snail gene encodes a key regulator of the epithelial-mesenchymal transition. Mol. Cell. Biol. 21:8184–8188.
  J5 P, ]# t) G/ x: n* v$ n
7 B9 x) p3 V1 I# |, ?/ ]4 ?Coghlan, M.P., A.A. Culbert, D.A. Cross, S.L. Corcoran, J.W. Yates, N.J. Pearce, O.L. Rausch, G.J. Murphy, P.S. Carter, L. Roxbee Cox, et al. 2000. Selective small molecule inhibitors of glycogen synthase kinase-3 modulate glycogen metabolism and gene transcription. Chem. Biol. 7:793–803.
' P( ^+ u+ b! A6 t% f1 S& I* V% X, f: b" z9 ?) `/ \
Diehl, J.A., M. Cheng, M.F. Roussel, and C.J. Sherr. 1998. Glycogen synthase kinase-3? regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 12:3499–3511." E2 z3 p5 A( N3 p+ A. D) q% p
! e2 D2 I8 Y( b7 z: v* t
Dom赤nguez, D., B. Montserrat-Sent赤s, A. Virg車s-Soler, S. Guaita, J. Grueso, M. Porta, I. Puig, J. Baulida, C. Franc赤, and A. Garc赤a de Herreros. 2003. Phosphorylation regulates the subcellular location and activity of the snail transcriptional repressor. Mol. Cell. Biol. 23:5078–5089.) R( T0 D3 `, h( P

: y( U% B6 v" y  x4 Q. l. V0 pGilles, C., M. Polette, M. Mestdagt, B. Nawrocki-Raby, P. Ruggeru, P. Birembaut, and J.M. Foidart. 2003. Transactivation of vimentin by ?-catenin in human breast cancer cells. Cancer Res. 63:2658–2664.4 b% R, I; O6 T4 Z( ^

# |3 y. W) j" m0 Y  n( A/ j, |5 GGrille, S.J., A. Bellacosa, J. Upson, A.J. Klein-Szanto, A.F. van Roy, W. Lee-Kwon, M. Donowitz, P.N. Tsichlis, and L. Larue. 2003. The protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 63:2172–2178.' ]+ Q, f/ Q" D6 g" l
) Q3 Y, E. i" n. y, H, j' c
Hirohashi, S. 1998. Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am. J. Pathol. 153:333–339.8 n+ [$ k6 h* }! z/ e

, J2 [8 q! F1 n' ^: V9 I  C/ DHuber, M.A., N. Azoitei, B. Baumann, S. Grunert, A. Sommer, H. Pehamberger, N. Kraut, H. Beug, and T. Wirth. 2004. NF-B is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J. Clin. Invest. 114:569–581.
" B! E9 r$ M1 @) U5 Q# }  T9 s2 R0 u- A5 J
Kim, K., Z. Lu, and E.D. Hay. 2002. Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol. Int. 26:463–476.
5 J# C* x$ z+ b& L0 S' Z( g" S2 \. Z$ C& i8 S( P
Kowalski, P.J., M.A. Rubin, and C.G. Kleer. 2003. E-cadherin expression in primary carcinomas of the breast and its distant metastases. Breast Cancer Res. 5:R217–R222.9 ^( a* }6 j: f8 h$ [' n
  b; z/ G1 [: {) Y
Liebner, S., A. Cattelino, R. Gallini, N. Rudini, M. Iurlaro, S. Piccolo, and E. Dejana. 2004. ?-Catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J. Cell Biol. 166:359–367.
, V, U: A2 G1 o( y
: i8 ?( h5 g5 z5 H/ Z" jMurray, N.R., L.A. Davidson, R.S. Chapkin, W. Clay Gustafson, D.G. Schattenberg, and A.P. Fields. 1999. Overexpression of protein kinase C ?II induces colonic hyperproliferation and increased sensitivity to colon carcinogenesis. J. Cell Biol. 145:699–711.
$ Y2 J7 m/ V6 ?/ a7 e; s' j& `! X; ]
Nelson, W.J., and R. Nusse. 2004. Convergence of Wnt, ?-catenin, and cadherin pathways. Science. 303:1483–1487.
$ L; H, r7 _9 s! M' z
4 ^" J/ ?5 x/ }6 \0 x: M0 E7 sOloumi, A., T. McPhee, and S. Dedhar. 2004. Regulation of E-cadherin expression and ?-catenin/Tcf transcriptional activity by the integrin-linked kinase. Biochim. Biophys. Acta. 1691:1–15.
0 I0 O9 b  X: N+ d9 \4 W0 n
3 S5 r/ f# a: T8 I: p! G3 q8 S! }Papkoff, J., and M. Aikawa. 1998. WNT-1 and HGF regulate GSK3? activity and ?-catenin signaling in mammary epithelial cells. Biochem. Biophys. Res. Commun. 247:851–855.' {( J$ N0 N/ O! e
  u8 X: B: S) Z* j' J
Peinado, H., M. Quintanilla, and A. Cano. 2003. Transforming growth factor ?-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 278:21113–21123.4 K: K2 I1 C6 H
, U* K" U. a8 w9 J; ~: u" _
Thiery, J.P. 2002. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer. 2:442–454.' P, z! q8 q. L" K) g

- [( X* M6 h- D* _6 N6 V. qWoodgett, J.R. 2001. Judging a protein by more than its name: GSK-3. Sci. STKE 2001:RE12.9 m# c# @; m$ k$ k

0 C2 K' N2 C' F+ e! f* {Zhou, B.P., J. Deng, W. Xia, J. Xu, Y.M. Li, M. Gunduz, and M.C. Hung. 2004. Dual regulation of Snail by GSK-3?-mediated phosphorylation in control of epithelial–mesenchymal transition. Nat. Cell Biol. 6:931–940./ b- z7 ]$ n. c/ j) Y
# z# e' p- y3 U5 s$ ]$ |
This article has been cited by other articles: (Search Google Scholar for Other Citing Articles)% r' c9 h: X  F! d& t- m& F2 d

- H1 g! v7 n6 L. c2 y: A; x/ E, yMartinez, M. J., Smith, A. D., Li, B., Zhang, M. Q., Harrod, K. S. (2007). Computational prediction of novel components of lung transcriptional networks. Bioinformatics 23: 21-29
% V& S- P/ o- f2 l* x" o, E
; D8 u7 K8 |+ @- J0 pLim, H.-Y., Tomlinson, A. (2006). Organization of the peripheral fly eye: the roles of Snail family transcription factors in peripheral retinal apoptosis. Development 133: 3529-3537
* t4 u$ x% u. B
+ u* m9 S( E' W0 x" _7 XDasari, V., Gallup, M., Lemjabbar, H., Maltseva, I., McNamara, N. (2006). Epithelial-Mesenchymal Transition in Lung Cancer: Is Tobacco the "Smoking Gun"?. Am. J. Respir. Cell Mol. Bio. 35: 3-97 \! m( U: d" c+ {

, X& K" I  e3 w1 V6 bRukstalis, J. M., Ubeda, M., Johnson, M. V., Habener, J. F. (2006). Transcription Factor Snail Modulates Hormone Expression in Established Endocrine Pancreatic Cell Lines. Endocrinology 147: 2997-3006
) P, A6 ^2 s; P& j$ k1 B5 u) n5 ?7 l* |4 h$ y) h
Medici, D., Hay, E. D., Goodenough, D. A. (2006). Cooperation between Snail and LEF-1 Transcription Factors Is Essential for TGF-beta1-induced Epithelial-Mesenchymal Transition. Mol. Biol. Cell 17: 1871-1879  f6 g& q; e( R' d/ ?' w8 Z8 q- h; T
) N# g6 j; ~2 N5 W6 G( E
Shin, S. R., Sanchez-Velar, N., Sherr, D. H., Sonenshein, G. E. (2006). 7,12-Dimethylbenz(a)Anthracene Treatment of a c-rel Mouse Mammary Tumor Cell Line Induces Epithelial to Mesenchymal Transition via Activation of Nuclear Factor-{kappa}B.. Cancer Res. 66: 2570-25755 _" l5 T4 S" W2 o  k/ B. ]2 a* f) u

& H' x& W) |* WIzzo, J. G., Malhotra, U., Wu, T.-T., Ensor, J., Luthra, R., Lee, J. H., Swisher, S. G., Liao, Z., Chao, K.S. C., Hittelman, W. N., Aggarwal, B. B., Ajani, J. A. (2006). Association of Activated Transcription Factor Nuclear Factor {kappa}B With Chemoradiation Resistance and Poor Outcome in Esophageal Carcinoma. J. Clin. Oncol. 24: 748-754
% n7 V$ N# E( d: C; q  X0 v
1 J8 {/ S: u1 i% m' lPeiro, S., Escriva, M., Puig, I., Barbera, M. J., Dave, N., Herranz, N., Larriba, M. J., Takkunen, M., Franci, C., Munoz, A., Virtanen, I., Baulida, J., de Herreros, A. G. (2006). Snail1 transcriptional repressor binds to its own promoter and controls its expression.. Nucleic Acids Res 34: 2077-2084
  e* @8 O- y+ p1 d8 h  J
- ^+ S) s% ^: OPlante, I., Cyr, D. G., Charbonneau, M. (2005). Involvement of the Integrin-Linked Kinase Pathway in Hexachlorobenzene-Induced Gender-Specific Rat Hepatocarcinogenesis. Toxicol Sci 88: 346-357
' I' `) A) H: H% [7 |, Q! e$ B* V6 `; Q
6 Y% S5 a0 l- a( f: R9 N( FSteinbrecher, K. A., Wilson, W. III, Cogswell, P. C., Baldwin, A. S. (2005). Glycogen Synthase Kinase 3{beta} Functions To Specify Gene-Specific, NF-{kappa}B-Dependent Transcription. Mol. Cell. Biol. 25: 8444-84556 G2 L9 p! v* p" L
& y/ v5 P* Z- E# ~8 V/ d  R
Carrozzino, F., Soulie, P., Huber, D., Mensi, N., Orci, L., Cano, A., Feraille, E., Montesano, R. (2005). Inducible expression of Snail selectively increases paracellular ion permeability and differentially modulates tight junction proteins. Am. J. Physiol. Cell Physiol. 289: C1002-C1014$ h- f, l% `6 b$ A
/ Z( H, z8 B3 D) @8 J# x1 C
Barrallo-Gimeno, A., Nieto, M. A. (2005). The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development 132: 3151-3161( f1 R" v% J$ R5 q8 K

! n8 A- t# u7 Y/ n1 bYoon, S.-O., Shin, S., Mercurio, A. M. (2005). Hypoxia Stimulates Carcinoma Invasion by Stabilizing Microtubules and Promoting the Rab11 Trafficking of the {alpha}6{beta}4 Integrin. Cancer Res. 65: 2761-2769(Robin E. Bachelder1, Sang-Oh Yoon1, Clar)

Rank: 2

积分
101 
威望
101  
包包
1951  
沙发
发表于 2015-6-7 10:58 |只看该作者
免疫细胞治疗  

Rank: 2

积分
72 
威望
72  
包包
1859  
藤椅
发表于 2015-6-25 10:42 |只看该作者
顶下再看  

Rank: 2

积分
107 
威望
107  
包包
1889  
板凳
发表于 2015-9-29 18:25 |只看该作者
干细胞之家微信公众号
不错,看看。  

Rank: 2

积分
64 
威望
64  
包包
1769  
报纸
发表于 2015-11-19 19:10 |只看该作者
先看看怎么样!  

Rank: 2

积分
132 
威望
132  
包包
1727  
地板
发表于 2015-11-26 20:07 |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
89 
威望
89  
包包
1794  
7
发表于 2015-12-14 16:08 |只看该作者
努力,努力,再努力!!!!!!!!!!!  

Rank: 2

积分
98 
威望
98  
包包
1756  
8
发表于 2015-12-17 08:10 |只看该作者
不是吧  

Rank: 2

积分
162 
威望
162  
包包
1724  
9
发表于 2016-1-12 17:58 |只看该作者
干细胞与基因技术

Rank: 2

积分
105 
威望
105  
包包
1851  
10
发表于 2016-2-15 20:43 |只看该作者
我帮你 喝喝  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-4-20 16:27

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.