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: 351935|回复: 264
go

RPTP is essential for NCAM-mediated p59fyn activation and neurite elon [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
3465  
楼主
发表于 2009-3-6 08:42 |只看该作者 |倒序浏览 |打印
1 Zentrum für Molekulare Neurobiologie, Universit?t Hamburg, 20246 Hamburg, Germany
) ]2 f8 E- e' Z% V1 v$ }/ y' l
  C- s0 H! L3 a. P/ b+ V$ b2 Hubrecht Laboratory, Netherlands Institute for Developmental Biology, 3584 CT Utrecht, Netherlands
3 y3 Y& P" ^8 f  n3 r. ~% W* m+ h# T+ v
Correspondence to Melitta Schachner: melitta.schachner@zmnh.uni-hamburg.de3 {7 U8 m$ t/ `3 e: f
/ x& H. N$ M2 T) _: ?$ @/ f7 f, c6 _
The neural cell adhesion molecule (NCAM) forms a complex with p59fyn kinase and activates it via a mechanism that has remained unknown. We show that the NCAM140 isoform directly interacts with the intracellular domain of the receptor-like protein tyrosine phosphatase RPTP, a known activator of p59fyn. Whereas this direct interaction is Ca2  independent, formation of the complex is enhanced by Ca2 -dependent spectrin cytoskeleton–mediated cross-linking of NCAM and RPTP in response to NCAM activation and is accompanied by redistribution of the complex to lipid rafts. Association between NCAM and p59fyn is lost in RPTP-deficient brains and is disrupted by dominant-negative RPTP mutants, demonstrating that RPTP is a link between NCAM and p59fyn. NCAM-mediated p59fyn activation is abolished in RPTP-deficient neurons, and disruption of the NCAM–p59fyn complex in RPTP-deficient neurons or with dominant-negative RPTP mutants blocks NCAM-dependent neurite outgrowth, implicating RPTP as a major phosphatase involved in NCAM-mediated signaling.
; g( ?4 l6 V) t
$ O' f: D$ Y6 O  h( eV. Bodrikov, I. Leshchyns'ka, and V. Sytnyk contributed equally to this paper." s7 d& c% D% l, _

4 G. @5 G; w+ g2 e4 HAbbreviations used in this paper: FGFR, FGF receptor; NCAM, neural cell adhesion molecule.
  A6 Z8 b9 Q9 y7 C
* J7 n, S1 s  T4 ^Introduction
- b% |) L1 W1 l: q; K2 m9 F' @' g
. ?9 |7 x2 R  |3 B2 fThe neural cell adhesion molecule (NCAM) is involved in several morphogenetic events, such as neuronal migration and differentiation, neurite outgrowth, and axon fasciculation. NCAM-induced morphogenetic effects depend on activation of Src family tyrosine kinases and, in particular, p59fyn kinase (Schmid et al., 1999). NCAM-dependent neurite outgrowth is impaired in neurons from p59fyn-deficient mice (Beggs et al., 1994) and is abolished by inhibitors of Src kinase family members (Crossin and Krushel, 2000; Kolkova et al., 2000; Cavallaro et al., 2001). The NCAM140 isoform has been observed in a complex with p59fyn, whereas p59fyn does not associate significantly with NCAM180 or glycosylphosphatidylinositol-linked NCAM120 (Beggs et al., 1997). However in oligodendrocytes, p59fyn is also associated with NCAM120 in isolated lipid rafts (Kramer et al., 1999), whereas in tumor cells NCAM is also associated with pp60c-src (Cavallaro et al., 2001), suggesting that additional molecular mechanisms may define NCAM's specificity of interactions with Src kinase family members. Several lines of evidence suggest that NCAM's association with lipid rafts is critical for p59fyn activation. NCAM not only colocalizes with p59fyn in lipid rafts (He and Meiri, 2002) but disruption of NCAM140 association with lipid rafts either by mutation of NCAM140 palmitoylation sites or by lipid raft destruction attenuates activation of the p59fyn kinase pathway, completely blocking neurite outgrowth (Niethammer et al., 2002). However, in spite of compelling evidence that NCAM can activate Src family tyrosine kinases, the mechanism of this activation has remained unclear.& m" e; h  \; d; R  r9 x) H
. a+ E: x8 @; y9 a: C8 V2 C) W! a
The activity of Src family tyrosine kinases is regulated by phosphorylation (Brown and Cooper, 1996; Thomas and Brugge, 1997; Bhandari et al., 1998; Hubbard, 1999; Petrone and Sap, 2000). The two best-characterized tyrosine phosphorylation sites in Src family tyrosine kinases perform opposing regulatory functions. The site within the enzyme's activation loop (Tyr-420 in p59fyn) undergoes autophosphorylation, which is crucial for achieving full kinase activity. In contrast, phosphorylation of the COOH-terminal site (Tyr-531 in p59fyn) inhibits kinase activity through intramolecular interaction between phosphorylated Tyr-531 and the SH2 domain in p59fyn, which stabilizes a noncatalytic conformation.
9 u. n* j% u& e5 V9 |5 R
: h8 E# D% j9 l# \& QA well known activator of Src family tyrosine kinases is the receptor protein tyrosine phosphatase RPTP (Zheng et al., 1992, 2000; den Hertog et al., 1993; Su et al., 1996; Ponniah et al., 1999). It contains two cytoplasmic catalytic domains, D1 and D2, of which only D1 is significantly active in vitro and in vivo (Wang and Pallen, 1991; den Hertog et al., 1993; Wu et al., 1997; Harder et al., 1998). To activate Src family tyrosine kinase, constitutively phosphorylated pTyr789 at the COOH-terminal of RPTP binds the SH2 domain of Src family tyrosine kinase that disrupts the intra-molecular association between the SH2 and SH1 domains of the kinase. This initial binding is followed by binding between the inhibitory COOH-terminal phosphorylation site of the Src family tyrosine kinase (pTyr531 in p59fyn) and the D1 domain of RPTP resulting in dephosphorylation of the inhibitory COOH-terminal phosphorylation sites in Src family tyrosine kinases (Zheng et al., 2000). These sites are hyperphosphorylated in cells lacking RPTP, and kinase activity of pp60c-src and p59fyn in RPTP-deficient mice is reduced (Ponniah et al., 1999). Like p59fyn and NCAM, RPTP is particularly abundant in the brain (Kaplan et al., 1990; Krueger et al., 1990), accumulates in growth cones (Helmke et al., 1998), and is involved in neural cell migration and neurite outgrowth (Su et al., 1996; Yang et al., 2002; Petrone et al., 2003).4 S$ K6 }, ~1 a% {7 |5 B
4 ?9 G" E- F" e0 ?6 ~8 W# u$ _! F
Remarkably, a close homologue of RPTP, CD45, associates with the membrane-cytoskeleton linker protein spectrin (Lokeshwar and Bourguignon, 1992; Iida et al., 1994), a binding partner of NCAM (Leshchyns'ka et al., 2003). Following this lead, we investigated the possibility that RPTP is involved in NCAM-induced p59fyn activation. We show that the intracellular domains of NCAM140 and RPTP interact directly and that this interaction is enhanced by spectrin-mediated Ca2 -dependent cross-linking of NCAM and RPTP. Levels of p59fyn associated with NCAM correlate with the ability of NCAM-associated RPTP to bind to p59fyn, and the NCAM–p59fyn complex is disrupted in RPTP-deficient brains implicating RPTP as linker molecule between NCAM and p59fyn. RPTP redistributes to lipid rafts in response to NCAM activation and RPTP levels are reduced in lipid rafts from NCAM-deficient mice, suggesting that NCAM recruits RPTP to lipid rafts to activate p59fyn. Finally, NCAM-mediated p59fyn activation is abolished in RPTP-deficient neurons and NCAM-induced neurite outgrowth is blocked in RPTP-deficient neurons or neurons transfected with dominant-negative RPTP mutants, demonstrating that RPTP is a major phosphatase involved in NCAM-mediated signaling.
: V! n6 p  K, z. U. Q
9 G* ~* b' {. ?' R% uResults
8 c- X9 X) p- M3 }+ V7 f9 I$ A7 A) v" {4 }  U, R8 V
Activation of p59fyn is impaired in NCAM-deficient brains
! r- q( O; `3 ]  O. B/ M# S7 v, y. Y, U5 p! o
Cross-linking of NCAM at the cell surface results in a rapid activation of p59fyn kinase (Beggs et al., 1997; Niethammer et al., 2002) via an unknown mechanism. To analyze whether or not NCAM deficiency may affect the activation status of p59fyn, we compared levels of activated p59fyn characterized by dephosphorylation at Tyr-531 and phosphorylation at Tyr-420 in the brains of wild-type and NCAM-deficient mice. Whereas the level of p59fyn protein was higher in brain homogenates of NCAM-deficient mice (Fig. 1 A), labeling with antibodies recognizing only p59fyn dephosphorylated at Tyr-531 or with antibodies recognizing only p59fyn phosphorylated at Tyr-420 was reduced in brain homogenates of NCAM-deficient mice (Fig. 1 B), indicating that activation of p59fyn is inhibited in NCAM-deficient brains and suggesting that NCAM is involved in the regulation of p59fyn function.4 D. @- f, }1 {3 ~2 g
! V. E, Q, [9 b. R
Figure 1. Activated p59fyn is reduced in NCAM-deficient brain. (A) Brain homogenates from 0- to 4-d-old wild-type (NCAM / ) and NCAM-deficient (NCAM–/–) mice were probed by Western blot with antibodies against total p59fyn protein. Labeling for GAPDH was included as loading control. Levels of p59fyn protein are increased in NCAM-deficient brains. (B) p59fyn immunoprecipitates from 0- to 4-d-old wild-type (NCAM / ) and NCAM-deficient (NCAM–/–) mice were probed by Western blot with antibodies against total p59fyn protein, p59fyn dephosphorylated at Tyr-531, or p59fyn phosphorylated at Tyr-420. Activated p59fyn is reduced in NCAM-deficient brains. Histograms (A and B) show quantitation of the blots with OD for wild type set to 100%. Mean values ± SEM (n = 6) are shown. *, P " _. q7 j8 Z5 v3 z7 T3 d0 G1 Q
6 t( a- q) f  I: h% w
NCAM forms a complex with RPTP! p. r4 H+ K+ w" q4 V2 B  l
! u( X% Y& n' D
The intracellular domain of NCAM does not contain sequences known to induce p59fyn activation. Thus, NCAM may form a complex with a protein, possibly a protein tyrosine phosphatase, to activate p59fyn. One possible candidate is the RPTP that dephosphorylates Tyr-531 of p59fyn (Bhandari et al., 1998) and is highly enriched in neurons and growth cones (Helmke et al.,1998). Remarkably, in RPTP-deficient cells, both dephosphorylation of the COOH-terminal tyrosine residue and autophosphorylation of the tyrosine residue within the activation loop of pp60c-src is reduced (von Wichert et al., 2003), resembling the phenotype of NCAM-deficient mice. Furthermore, a close homologue of RPTP, CD45, associates with the membrane-cytoskeleton linker protein spectrin (Lokeshwar and Bourguignon, 1992; Iida et al., 1994), a binding partner of NCAM (Leshchyns'ka et al., 2003). To investigate if NCAM interacts with RPTP, we analyzed the distribution of both proteins in cultured hippocampal neurons. NCAM and RPTP partially colocalized along neurites, and both proteins accumulated in growth cones where clusters of NCAM partially overlapped with accumulations of RPTP (Fig. 2 A). To verify whether or not NCAM interacts with RPTP, we induced clustering of NCAM at the cell surface of live hippocampal neurons by incubation with antibodies against NCAM. Clustering of NCAM enhanced overlap between NCAM and RPTP localization (mean correlation between NCAM and RPTP localization being 0.3 ± 0.05 and 0.6 ± 0.03 in neurons treated with nonspecific IgG or NCAM antibodies, respectively; Fig. 2 B), indicating that RPTP partially redistributed to NCAM clusters and suggesting that NCAM and RPTP form a complex. Because antibodies against RPTP were directed against its intracellular domain, RPTP contained in intracellular organelles could have been recognized as colocalizing with NCAM that associates with intracellular organelles of trans-Golgi network origin (Sytnyk et al., 2002). Thus, the redistribution of RPTP to NCAM clusters may represent redistribution of intracellular carriers containing RPTP. To analyze whether or not NCAM associates with RPTP in the plasma membrane, we transfected neurons with RPTP containing the HA tag in the extracellular domain and induced clustering of NCAM and HA-RPTP with antibodies against NCAM and the HA tag. HA-RPTP partially redistributed to NCAM clusters (Fig. 2 C), indicating that both proteins form a complex at the cell surface.
' ~# A+ j& U; {) Y9 t0 t; @+ h3 P8 k  [( Q6 E' v4 @7 H
Figure 2. NCAM forms a complex with RPTP. (A) High magnification image of a growth cone of a hippocampal neuron labeled with antibodies against NCAM and RPTP. Note that clusters of NCAM overlap with accumulations of RPTP. (B) Live hippocampal neurons were treated with nonspecific IgG or with antibodies against NCAM. Note that antibodies against NCAM induced clustering of NCAM at the cell surface. Labeling with antibodies against RPTP showed that RPTP partially redistributed to NCAM clusters (arrows). The corresponding profiles show NCAM and RPTP labeling intensities along neurites. Note increased overlap of NCAM and RPTP clusters in neurons treated with NCAM antibodies. (C) Hippocampal neurons transfected with wild-type RPTP containing an HA tag extracellularly were incubated live with antibodies against the HA tag and NCAM. Cell surface RPTP partially redistributed to NCAM clusters (arrows). Bars, 10 μm. (D) Brain homogenates of wild-type (NCAM / ) and NCAM-deficient (NCAM–/–) mice (total brain) and NCAM immunoprecipitates (IP: NCAM) were probed with antibodies against RPTP by Western blot. Labeling for GAPDH was included as loading control. RPTP coimmunoprecipitates with NCAM. Note increased expression of RPTP in NCAM-deficient brains. Histogram shows quantitation of the RPTP level in wild type ( / ) and NCAM-deficient (–/–) brains. OD for wild type was set to 100%. Mean values ± SEM (n = 6) are shown. *, P
1 l: v. m4 l4 C" t) Z; U) r, ~) |7 Z* O9 @0 Z, I8 A2 @
Finally, we examined the association between NCAM and RPTP in the brain by coimmunoprecipitation. RPTP coimmunoprecipitated with NCAM from brain homogenates (Fig. 2 D), confirming that NCAM associates with RPTP. Interestingly, we found that the level of RPTP was approximately two times higher in the brain of NCAM-deficient mice when compared with wild-type mice (Fig. 2 D), indicating a functional relationship between NCAM and RPTP.
+ c' g* h5 R( y) F3 D$ A- F7 v& d9 @
7 X4 K; e7 {) H1 t1 oNCAM140 is the most potent RPTP-binding NCAM isoform1 ~) {0 x2 c7 }' [" I$ x% x8 o
8 N6 d, ?, g4 H% E. M+ E! r
To identify the NCAM isoform interacting with RPTP, we expressed NCAM120, NCAM140, and NCAM180 in CHO cells and analyzed their association with RPTP by coimmunoprecipitation. CHO cells endogenously express RPTP that was detected with RPTP antibodies as a band with a molecular mass identical to RPTP detected in brain homogenates (unpublished data). Although transfected CHO cells expressed NCAM120, NCAM140, and NCAM180 in similar amounts, RPTP coimmunoprecipitated only with NCAM140 (Fig. 3 A). However, after prolonged exposure of the film we could also detect RPTP in NCAM180 immunoprecipitates (unpublished data). RPTP did not coimmunoprecipitate with NCAM120. We conclude that RPTP associates predominantly with NCAM140 and to a lesser extent with NCAM180.: N9 |' M# w1 l6 d" P2 |

* C8 Q) d& c6 e7 DFigure 3. Intracellular domain of NCAM140 directly interacts with the intracellular domain of RPTP. (A) Lysates and NCAM immunoprecipitates (IP: NCAM) from CHO cells transfected with NCAM120, NCAM140, NCAM180, or GFP were probed by Western blot with antibodies against NCAM and RPTP. Note that NCAM isoforms were expressed in approximately equal amounts whereas RPTP immunoprecipitated only with NCAM140 but not NCAM180 or NCAM120. Labeling for GAPDH was included as loading control. (B) Intracellular domains of NCAM140, NCAM180, or CHL1 were bound to Ni-NTA agarose beads. The gel was stained with Coomassie blue and shows that approximately equal amounts of the intracellular domains of NCAM140, NCAM180, or CHL1 were bound to beads. Beads were incubated with equal concentrations of intracellular domains of RPTP and the extent of RPTP intracellular domain binding was determined by Western blotting using polyclonal antibodies against RPTP. Intracellular domains of RPTP interacted with intracellular domains of NCAM140, and to a lesser extent NCAM180, but not with intracellular domains of CHL1. (C) Intracellular domains of NCAM140, NCAM180, or CHL1 were bound to plastic and assayed by ELISA for the ability to bind increasing concentrations of RPTP intracellular domains. Binding to BSA served as a control. Mean values (OD 405) ± SEM (n = 6) are shown. Intracellular domains of RPTP bound to intracellular domains of NCAM140 and with a lower affinity to intracellular domains of NCAM180, but not to intracellular domains of CHL1.( \  e) u! z+ o8 K
- D( U5 g& J& N+ D+ L9 f
Inability of NCAM120, the GPI-linked NCAM isoform without the intracellular domain, to bind RPTP suggested that the intracellular domain of NCAM is involved in the formation of a complex between NCAM and RPTP. Furthermore, the extracellular domain of NCAM (NCAM-Fc) did not bind to RPTP in brain lysates, confirming that the extracellular domain of NCAM does not bind to RPTP (unpublished data). To verify that the NCAM intracellular domain interacts directly with the intracellular domain of RPTP, we analyzed binding of the recombinant intracellular domain of RPTP to the intracellular domain of NCAM180 or NCAM140 in a pull-down assay. For comparison, the intracellular domain of CHL1, another adhesion molecule of the immunoglobulin superfamily, was used. The intracellular domain of RPTP bound to the intracellular domain of NCAM180 or NCAM140 but not to the intracellular domain of CHL1 (Fig. 3 B). Interaction between the intracellular domains of RPTP and NCAM140 was severalfold stronger than between the intracellular domains of RPTP and NCAM180 (Fig. 3 B). To confirm this finding, we examined the direct interaction between the intracellular domains of RPTP and NCAM180 or NCAM140 by ELISA. Intracellular domain of RPTP bound to the intracellular domains of NCAM180 or NCAM140 in a concentration-dependent manner, with the intracellular domain of NCAM140 binding with a higher affinity than the intracellular domain of NCAM180 (Fig. 3 C). No binding with the intracellular domain of CHL1 was observed (Fig. 3 C). We conclude that NCAM binds directly to RPTP via the intracellular domain, with NCAM140 being the most potent RPTP-binding NCAM isoform.
$ Z6 h1 `! e6 n' c& H9 x6 A5 ?# _( G: h+ I! C- N
RPTP binds NCAM140 via the D2 domain and links NCAM140 to p59fyn
  k! o* K$ j) w1 Y6 |2 o8 m" H4 b' I. M  m: X# z" B0 K# a
To identify the part of the intracellular domain of RPTP responsible for the interaction with NCAM140, we coexpressed, in CHO cells, NCAM140 together with the wild-type form of RPTP (wtRPTP), RPTP lacking the D2 domain (RPTPD2), or catalytically inactive form of RPTP containing a mutation within the D1 catalytic domain (RPTPC433S) and analyzed binding of NCAM140 to these RPTP mutants by coimmunoprecipitation. All transfected RPTP constructs contained the HA tag to distinguish them from endogenous RPTP. As seen for endogenous RPTP, transfected wtRPTP coimmunoprecipitated with NCAM140 (Fig. 4 A). Similar amounts of RPTPC433S coimmunoprecipitated with NCAM140, whereas RPTPD2 did not coimmunoprecipitate (Fig. 4 A), indicating that the D2 domain is required for the interaction between RPTP and NCAM140.
& ?% Z: B4 T4 Q, }% _
+ n4 O7 k, D5 ~4 h3 B" }Figure 4. RPTP interacts with NCAM140 via D2 domain and links NCAM140 to p59fyn. (A) Lysates from CHO cells cotransfected with NCAM140 and wild-type (wt) RPTP, RPTPD2, RPTPC433S, or GFP were probed with antibodies against HA tag and NCAM by Western blot. NCAM140 and RPTP constructs are expressed at approximately equal amounts in transfected CHO cells. Labeling for GAPDH was included as loading control. NCAM (IP: NCAM) or RPTP (IP: HA) immunoprecipitates were probed with antibodies against HA tag and p59fyn. wtRPTP and RPTPC433S but not RPTPD2 coimmunoprecipitated with NCAM140. RPTPC433S and RPTPD2 inhibited coimmunoprecipitation of p59fyn with NCAM140. p59fyn coimmunoprecipitated with wtRPTP and RPTPD2 but to a lower extent with RPTPC433S. (B) Hippocampal neurons transfected with RPTPC433S, RPTPD2, or GFP were incubated live with NCAM antibodies to cluster NCAM and fixed and labeled with antibodies against p59fyn. Note that redistribution of p59fyn to NCAM clusters (arrows) was reduced in neurons transfected with RPTPC433S or RPTPD2. Bar, 10 μm. The corresponding profiles show NCAM and p59fyn labeling intensities along transfected neurites. The histogram shows mean labeling intensity of p59fyn in NCAM clusters. Transfection with RPTPC433S or RPTPD2 reduced association between p59fyn and NCAM. Mean values ± SEM (n > 20 neurons) are shown in arbitrary units (AU). *, P . q7 C! y3 e8 Z6 U# ^- ]5 n5 ~

- ^5 H7 j& r0 ^' t, HRemarkably, among the major NCAM isoforms, only NCAM140 forms a complex with p59fyn (Beggs et al., 1997) that we found to correlate with its ability to bind RPTP (see the previous section). RPTP directly interacts with p59fyn (Bhandari et al., 1998). Accordingly, p59fyn coimmunoprecipitated with wtRPTP from transfected CHO cells (Fig. 4 A). Approximately the same amount of p59fyn coimmunoprecipitated with RPTPD2 (Fig. 4 A), indicating that this truncated construct also binds p59fyn probably via the D1 domain. In accordance with previous reports, p59fyn showed reduced ability to bind RPTPC433S, a catalytically inactive mutant of RPTP (Fig. 4 A; Zheng et al., 2000).' F  ?6 W" v' s. c. [

' o4 P+ ~" \6 V, J  x  }To analyze the role of RPTP in NCAM140–p59fyn complex formation, we coimmunoprecipitated p59fyn with NCAM140 in the presence of RPTP mutants. In CHO cells cotransfected with NCAM140 and wtRPTP, p59fyn coimmunoprecipitated with NCAM140 (Fig. 4 A). The amount of p59fyn coimmunoprecipitated with NCAM140 was reduced in cells cotransfected with RPTPC433S (Fig. 4 A), correlating with the reduced ability of this catalytically inactive RPTP mutant to bind p59fyn (see previous paragraph; Zheng et al., 2000). When NCAM140 was cotransfected with RPTPD2, p59fyn no longer coimmunoprecipitated with NCAM140 (Fig. 4 A). Because RPTPD2 binds p59fyn (Fig. 4 A), it is conceivable that this mutant, which does not bind NCAM140, competes with endogenous RPTP for binding to p59fyn and thus inhibits NCAM140–p59fyn complex formation.
8 t4 M' F- l, A* X- A% s/ v' F
5 j! L0 A( f  ?" e5 Y6 Q6 C% oTo extend this analysis to neurons, we transfected hippocampal neurons with GFP alone or cotransfected with GFP and RPTPD2 or RPTPC433S and analyzed the redistribution of p59fyn to NCAM clusters after cross-linking NCAM with NCAM antibodies (Fig. 4 B). In neurons transfected with RPTPD2 or RPTPC433S, the level of p59fyn in NCAM clusters was reduced by 30% when compared with GFP only transfected cells, suggesting that RPTPD2 or RPTPC433S inhibit NCAM–p59fyn complex formation by competing with endogenous RPTP. The combined observations indicate that NCAM140–p59fyn complex formation correlates with the ability of NCAM140-associated RPTP to bind to p59fyn, implicating RPTP as a linker between NCAM140 and p59fyn.
3 L$ B, q2 T. h* J% D9 K) o9 y3 t9 S% x# i1 A: [2 j) Y4 B
Association between NCAM and p59fyn and NCAM-mediated p59fyn activation are abolished in RPTP-deficient neurons0 }2 e0 Z, x. t8 B

& l. D* E& ?) A6 A" p8 D* xTo substantiate further our finding that RPTP is a linker protein between NCAM and p59fyn, we analyzed p59fyn activation and association of p59fyn with NCAM in RPTP-deficient brains. As for NCAM-deficient brains, levels of p59fyn dephosphorylated at Tyr-531 and levels of p59fyn phosphorylated at Tyr-420 were reduced in brain homogenates of RPTP-deficient mice (Fig. 5 A), further suggesting that RPTP plays a role in NCAM-mediated p59fyn activation in the brain. To analyze the role of RPTP in the formation of the complex between NCAM and p59fyn, we immunoprecipitated NCAM from wild-type and RPTP-deficient brains and probed immunoprecipitates with antibodies against p59fyn. Whereas p59fyn coimmunoprecipitated with NCAM from wild-type brains, p59fyn did not coimmunoprecipitate with NCAM from RPTP-deficient brains (Fig. 5 B). Furthermore, when NCAM was clustered at the surface of wild-type and RPTP-deficient cultured hippocampal neurons, levels of p59fyn were significantly reduced in NCAM clusters in RPTP-deficient neurons when compared with wild-type cells (Fig. 5 C), indicating that RPTP is required for complex formation between NCAM and p59fyn.
7 h. e5 d1 O: X8 R
( x7 y" q* W& g# P: z% h5 eFigure 5. NCAM–p59fyn complex formation and NCAM-mediated p59fyn activation are abolished in RPTP-deficient neurons. (A) p59fyn immunoprecipitates from 4-d-old wild-type (RPTP / ) and RPTP-deficient (RPTP–/–) brain homogenates were probed by Western blot with antibodies against total p59fyn protein, p59fyn dephosphorylated at Tyr-531, or p59fyn phosphorylated at Tyr-420. Levels of p59fyn dephosphorylated at Tyr-531 and p59fyn phosphorylated at Tyr-420 are reduced in RPTP-deficient brains. Histograms show quantitation of the blots with OD for wild type set to 100%. Mean values ± SEM (n = 6) are shown. *, P  20 neurons) are shown in arbitrary units (AU). *, P  20 neurons) are shown in arbitrary units (AU). *, P
- }  U! w* B& ~2 T$ k8 N, ]9 h/ ]7 h6 G' k: _" @
NCAM clustering at the cell surface induces rapid p59fyn activation (Beggs et al., 1997). To analyze whether or not RPTP is required for NCAM-induced p59fyn activation, we treated live hippocampal neurons from wild-type and RPTP-deficient mice with NCAM antibodies and analyzed levels of p59fyn dephosphorylated at Tyr-531 along neurites of the stimulated neurons. Clustering of NCAM increased levels of Tyr-531–dephosphorylated p59fyn along neurites of wild-type neurons by 60% (Fig. 5 D). However, NCAM-mediated p59fyn activation was completely abolished in RPTP-deficient neurons (Fig. 5 D), demonstrating that RPTP is required for NCAM-mediated p59fyn activation.
6 x: b# Q, }; J; B/ a' R3 z: c# a2 L/ l) g
Formation of the complex between RPTP and NCAM is enhanced by Ca2 + g9 ~. h' X2 A; N  |' w/ V

  n$ T9 J/ M: ~! z/ TCoimmunoprecipitation experiments were performed either in the presence of Ca2  or with 2 mM EDTA, a Ca2 -sequestering agent. Whereas RPTP coimmunoprecipitated with NCAM from brain homogenates under both conditions, coimmunoprecipitated complexes were reduced by 60% in the presence of EDTA (Fig. 6 A), suggesting that Ca2  promotes formation of the NCAM–RPTP complex. These results are in accordance with findings of Zeng et al. (1999), who found that NCAM and RPTP did not coimmunoprecipitate in the presence of EDTA. To analyze if the direct interaction between NCAM and RPTP is Ca2  dependent, we assayed binding of the intracellular domain of NCAM140 to the intracellular domain of RPTP by ELISA in the presence or absence of Ca2  (Fig. 6 B), showing that the direct interaction is Ca2  independent and suggesting that additional binding partners of NCAM and/or RPTP may enhance complex formation in a Ca2 -dependent manner. Spectrin, which directly interacts with the intracellular domain of NCAM (Leshchyns'ka et al., 2003) and contains a Ca2  binding domain (De Matteis and Morrow, 2000), is one of the possible candidates. Indeed, RPTP coimmunoprecipitated with spectrin from brain homogenates (Fig. 6 C). In the presence of 2 mM EDTA, RPTP coimmunoprecipitating with spectrin was reduced by 80% (Fig. 6 C), whereas coimmunoprecipitation of NCAM with spectrin did not depend on Ca2  (Fig. 6 C). We conclude that RPTP directly interacts with NCAM in a Ca2 -independent manner. However, formation of the complex is enhanced by Ca2 -dependent cross-linking of NCAM140 and RPTP via spectrin.0 ^2 ^6 R5 T3 s# p; |
' Q# Z# G: f0 }0 j6 y
Figure 6. NCAM–RPTP complex formation is enhanced by spectrin in a Ca2 -dependent manner. (A) NCAM immunoprecipitates obtained from brain homogenates of wild-type mice ( / ) in the presence or absence of 2 mM EDTA were probed by Western blot with antibodies against RPTP. NCAM-deficient brains (–/–) were taken for control. Coimmunoprecipitation of RPTP was inhibited by EDTA application. The histogram shows quantitation of the blots. (B) Intracellular domains of NCAM140 were bound to plastic and assayed by ELISA for the ability to bind increasing concentrations of RPTP intracellular domains in the absence of Ca2  or in the presence of 2 mM Ca2 . Binding to BSA served as a control. Mean values (OD 405) ± SEM (n = 6) are shown. Binding of intracellular domains of RPTP to intracellular domains of NCAM140 did not depend on Ca2 . (C) Spectrin immunoprecipitates obtained from brain homogenates of wild-type mice in the absence or presence of 2 mM EDTA were probed by Western blot with antibodies against RPTP or NCAM. Non-immune rabbit Ig (control Ig) were used for control. Coimmunoprecipitation of RPTP with spectrin was inhibited by EDTA, whereas coimmunoprecipitation of NCAM did not depend on Ca2 . Histograms show quantitation of the blots. For histograms in A and C, OD in the presence of Ca2  was set to 100% and mean values ± SEM (n = 6) are shown. *, P
) b  C" T8 J& u& R$ C. y! Y: t$ I6 |4 M, Q' g; ~1 U+ {
The NCAM–RPTP complex redistributes to lipid rafts after NCAM activation
6 S( e9 K# B% _( a7 Q- j# g4 @
8 e. C/ X* G5 _% z. @) m8 _Whereas p59fyn is mainly associated with lipid rafts (van't Hof and Resh, 1997; Niethammer et al., 2002; Filipp et al., 2003), only 4–8% of all RPTP molecules were found in lipid rafts of brain (unpublished data). In hippocampal neurons extracted with cold 1% Triton X-100 to isolate lipid rafts (Niethammer et al., 2002; Leshchyns'ka et al., 2003), detergent-insoluble clusters of RPTP only partially overlapped with the lipid raft marker ganglioside GM1 (Fig. 7 A), further confirming that RPTP and p59fyn are segregated at the subcellular level. Because activation of NCAM results in its redistribution to lipid rafts (Leshchyns'ka et al., 2003), it may also promote redistribution of NCAM-associated RPTP to lipid rafts and thus activate raft-associated p59fyn. To verify this hypothesis, we studied association of NCAM and RPTP with lipid rafts in hippocampal neurons activated or not activated with NCAM-Fc or NCAM antibodies. In accordance with previous results (Leshchyns'ka et al., 2003), application of NCAM-Fc or NCAM antibodies increased GM1 levels in detergent-insoluble clusters of NCAM, indicating that NCAM redistributed to lipid rafts (Fig. 7, A–C). Application of NCAM-Fc or NCAM antibodies also increased the level of RPTP in NCAM clusters, indicating that NCAM activation promoted NCAM–RPTP complex formation (Fig. 7, A–C). Furthermore, NCAM activation also increased GM1 levels in detergent-insoluble clusters of RPTP (Fig. 7 D), confirming that NCAM-associated RPTP also redistributed to lipid rafts and suggesting that NCAM recruits RPTP to lipid rafts. To further analyze this possibility, we compared levels of RPTP in lipid rafts in brains of wild-type and NCAM-deficient mice. Indeed, RPTP was reduced by 60% in lipid rafts isolated from NCAM-deficient brains (Fig. 7 E), confirming that NCAM plays a role in RPTP targeting to lipid rafts. The levels of p59fyn were increased in NCAM-deficient lipid rafts (100% and 124 ± 7.6% in wild-type and NCAM deficient rafts, respectively) probably reflecting increased levels of p59fyn in NCAM-deficient brains. Levels of GM1 were not different in lipid rafts from wild-type and NCAM-deficient brains (100% and 103.3 ± 6.8% in wild-type and NCAM-deficient rafts, respectively), showing that lipid rafts were isolated with the same efficacy from wild-type and NCAM-deficient brains (Fig. 7 E).4 }7 L9 v; q8 m( ]! N, _

" P* T$ L6 N5 Z7 q5 m6 LFigure 7. NCAM activation induces redistribution of RPTP to lipid rafts. (A–D and F) Control hippocampal neurons and neurons incubated with corresponding reagents were extracted in cold 1% Triton X-100 and labeled with antibodies against NCAM and RPTP together with FITC-labeled cholera toxin to visualize GM1-containing lipid rafts. (A) Note increased overlap of NCAM with RPTP and GM1 after NCAM-Fc application when compared with control neurons (arrows). Bar, 10 μm. (B and C) Histograms show mean intensities of RPTP and GM1 in NCAM clusters in control and NCAM-Fc (B) or NCAM antibody (C)–treated neurons. (D) Histograms show mean intensities of NCAM and GM1 in RPTP clusters in control and NCAM antibody–treated neurons. (E) Lipid rafts obtained from wild-type ( / ) or NCAM-deficient brains (–/–) were probed by Western blot (WB) with antibodies against RPTP or p59fyn or by dot blot (DB) with cholera toxin. Note reduced amount of RPTP in rafts from NCAM-deficient brains. The histogram shows quantitation of the RPTP levels in lipid rafts. OD in wild type was set to 100% and mean values ± SEM (n = 6) are shown. *, P  50) in arbitrary units (AU). *, P
- U$ I3 X& v3 m' \
7 B) Y- @7 H  f( Q! v, X( y' yNCAM-mediated recruitment of RPTP to lipid rafts is enhanced by NCAM-induced FGF receptor (FGFR)–dependent increase in intracellular Ca2
+ K3 o' ^* ?) ^5 y* P
* P+ w9 H8 `' z8 v) rNCAM activation increases intracellular Ca2  concentrations via a FGFR-dependent mechanism (Walsh and Doherty, 1997; Kamiguchi and Lemmon, 2000; Juliano, 2002). This increase in intracellular Ca2  may account for the enhanced association between NCAM and RPTP after NCAM activation (Fig. 7, A–D) because of spectrin-mediated cross-linking of NCAM140 and RPTP (Fig. 6). Interestingly, NCAM activation also induces redistribution of NCAM-associated spectrin to lipid rafts (Leshchyns'ka et al., 2003). To analyze the role of FGFR and Ca2  in the recruitment of RPTP to an NCAM complex, we estimated levels of RPTP associated with NCAM following NCAM activation in control neurons and neurons incubated with BAPTA-AM, a membrane-permeable Ca2  chelator (Williams et al., 1992; Cavallaro et al., 2001), or a specific FGFR inhibitor (Niethammer et al., 2002; Leshchyns'ka et al., 2003). Whereas NCAM activation increased levels of RPTP and GM1 in NCAM clusters (Fig. 7 F), treatment with BAPTA-AM or FGFR inhibitor abolished recruitment of RPTP to NCAM clusters in response to NCAM activation (Fig. 7 F). In accordance with previous findings (Leshchyns'ka et al., 2003), NCAM redistribution to lipid rafts was not affected by the FGFR inhibitor or BAPTA-AM (Fig. 7 F). BAPTA-AM or FGFR inhibitor did not affect the level of RPTP associated with NCAM under nonactivated conditions (Fig. 7 F). We conclude that, whereas at resting conditions Ca2  does not play a major role in the interaction between NCAM and RPTP, NCAM-induced FGFR-dependent elevations of intracellular Ca2  levels strengthen the interactions between NCAM and RPTP in response to NCAM activation, most likely via spectrin (see the section Formation of the complex between RPTP and NCAM is enhanced by Ca2 ).8 c1 P" g4 x) G5 S# k% }
4 D* R  {2 }% d, y4 d  s
NCAM-induced neurite outgrowth depends on NCAM association with RPTP( I$ R! O* g6 {! l+ t3 f
1 s: T/ i' V% E2 m' C0 [3 |
NCAM-induced neurite outgrowth depends on p59fyn activation (Kolkova et al., 2000), suggesting that NCAM association with RPTP may be involved. To analyze the role of protein tyrosine phosphatases in NCAM-induced neurite outgrowth, we incubated cultured hippocampal neurons with 100 μM vanadate, an inhibitor of these phosphatases (Helmke et al., 1998). NCAM-Fc–enhanced neurite outgrowth was abolished by vanadate, indicating that activation of protein tyrosine phosphatases is required for NCAM-mediated neurite outgrowth. Vanadate did not affect neurite outgrowth in nonstimulated neurons, indicating that vanadate does not lead to nonspecific impairments (Fig. 8 A). To directly assess the role of RPTP in NCAM-induced neurite outgrowth, we transfected hippocampal neurons with the dominant-negative mutants of RPTP. Both, RPTPD2, which does not bind NCAM but associates with p59fyn, and catalytically inactive RPTPC433S, which associates with NCAM but binds p59fyn with a lower efficiency than endogenous RPTP, inhibited association of NCAM with p59fyn by competing with endogenous RPTP (Fig. 4). In neurons transfected with GFP only, stimulation with NCAM-Fc significantly enhanced neurite length when compared with control nonstimulated neurons (Fig. 8 B). However, neurons transfected with RPTPD2 or RPTPC433S remained unresponsive to NCAM-Fc stimulation (Fig. 8 B), indicating that RPTP plays a major role in NCAM-induced neurite outgrowth. To confirm this finding, we analyzed NCAM-mediated neurite outgrowth in hippocampal neurons from RPTP-deficient mice. Whereas NCAM-Fc enhanced neurite outgrowth in neurons from RPTP wild-type littermates by 100%, NCAM-Fc–induced neurite outgrowth was completely abolished in RPTP-deficient neurons (Fig. 8 C), further confirming that RPTP is required for NCAM-mediated neurite outgrowth.
. v' f2 g+ v8 J" i1 ?; i. Z' B8 F6 c/ l- Y
Figure 8. NCAM-mediated neurite outgrowth depends on RPTP activation. (A) Hippocampal neurons were incubated with NCAM-Fc alone or with NCAM-Fc together with vanadate, and lengths of the longest neurites were measured. NCAM-Fc increased neurite length when compared with control neurons. Vanadate decreased neurite outgrowth in the NCAM-Fc–stimulated group to the control group level but did not affect basal neurite outgrowth over poly-L-lysine. (B) Hippocampal neurons transfected with GFP alone or cotransfected with GFP and RPTPC433S or RPTPD2 were incubated with NCAM-Fc after transfection and lengths of the longest neurites were measured. NCAM-Fc increased neurite lengths in GFP-transfected neurons. NCAM-Fc–stimulated neurite outgrowth was blocked in the group cotransfected with RPTPC433S or RPTPD2. (C) Lengths of the longest neurites were measured in wild-type ( / ) and RPTP-deficient (–/–) neurons not treated or treated with NCAM-Fc. NCAM-Fc increased neurite length in wild-type but not in RPTP-deficient neurons. For A–C, mean values ± SEM are shown (n > 150 neurons; *, P - F' b* ^8 S% D( {
8 C8 [" r7 y9 M# ^4 f
Discussion
9 i2 p# \/ U! i6 h4 Z9 d, k: H5 G. E- r: `
It is by now well established that in response to homophilic or heterophilic binding cell adhesion molecules of the immunoglobulin superfamily, such as NCAM, L1, or CHL1, activate Src family tyrosine kinases, and in particular pp60c-src or p59fyn, resulting in morphogenetic events, such as cell migration and neurite outgrowth. However, the mechanisms of Src family tyrosine kinase activation in these paradigms have remained unresolved. Here, we identify a cognate activator of p59fyn, the receptor protein tyrosine phosphatase RPTP, as a novel binding partner of NCAM. Activation of p59fyn is reduced in NCAM-deficient mice and interaction between NCAM and p59fyn is abolished in RPTP-deficient brains. Interestingly, we found that the levels of p59fyn and RPTP are increased in NCAM-deficient brains, possibly reflecting a compensatory reaction to the decreased activity of these enzymes in the mutant and further indicating a tight functional relationship between NCAM, RPTP, and p59fyn. NCAM-induced neurite outgrowth is completely abrogated in RPTP-deficient neurons or in neurons transfected with dominant-negative RPTP mutants, indicating that RPTP links NCAM to p59fyn both physically and functionally.
- v2 D/ R; t& O3 F2 E7 ^$ Z, H5 k
. A. Z3 m2 o- D1 f! zRole of Ca2  in NCAM–RPTP–p59fyn complex formation
  K, G9 z2 ]% q; c: |  h$ `0 S( h5 o7 F" K2 m, w" A8 k" S2 [! q
Interactions between NCAM and RPTP and NCAM–RPTP–p59fyn complex formation leading to neurite outgrowth are tightly regulated (Fig. 9). First, whereas direct interaction between NCAM and RPTP is Ca2  independent, NCAM–RPTP complex formation is enhanced by Ca2 -dependent cross-linking via spectrin. Remarkably, NCAM activation results in an increase in intracellular Ca2  concentration via influx through Ca2  channels or release from intracellular stores, and may thus provide a positive feedback loop between NCAM activation and NCAM–RPTP complex formation involving spectrin. RPTP binding to spectrin may also elevate RPTP enzymatic dephosphorylation activity (Lokeshwar and Bourguignon, 1992). Interestingly, NCAM activation also induces activation of PKC (Kolkova et al., 2000; Leshchyns'ka et al., 2003), which is known to phosphorylate RPTP and stimulate its activity (den Hertog et al., 1995; Tracy et al., 1995; Zheng et al., 2002). Thus, a network of activated intracellular signaling molecules may underlie the induction and maintenance of NCAM-mediated neurite outgrowth. It is interesting in this respect that the NCAM140 isoform predominates in these interactions: it interacts more efficiently with p59fyn via RPTP and enhances neurite outgrowth more vigorously than NCAM180 (Niethammer et al., 2002). The structural dispositions of NCAM140 for this preference will remain to be established.) j% z% i9 u  K3 l

  S& Z9 R: h4 n! S7 u9 }& XFigure 9. A proposed model of NCAM140-mediated p59fyn activation cascade. Before NCAM activation, NCAM140 and RPTP located predominantly in raft-free areas are segregated from p59fyn, which predominantly associates with lipid rafts. NCAM activation induces FGFR-dependent increase in intracellular Ca2  that enhances RPTP–NCAM140 complex formation via spectrin as a cross-linking platform. Additionally, NCAM activation results in the redistribution of the complex to lipid rafts due to NCAM palmitoylation. In lipid rafts, RPTP binds and dephosphorylates p59fyn resulting in p59fyn activation, which, in turn, promotes neurite outgrowth.
% `6 H1 T$ x; R5 ?; `) e5 r3 _+ L9 }1 [9 ^. a# E6 P5 X
The role of lipid rafts
) J6 @, n  v2 F; G( g% c% ]9 }# A/ `$ e9 t/ P9 [" z+ g3 q
Additional regulation of RPTP-mediated p59fyn activation is achieved by segregation of RPTP and p59fyn to different subdomains in the plasma membrane (Fig. 9). Approximately 90% of all RPTP molecules in the brain are located in a lipid raft-free environment and are thereby segregated from lipid raft-associated p59fyn under nonstimulated conditions. Whereas segregation of receptor protein tyrosine phosphatases from their potential substrates due to targeting to different plasma membrane domains has been suggested as a general mechanism of the regulation of receptor protein tyrosine phosphatase function (Petrone and Sap, 2000), the mechanisms that target receptor protein tyrosine phosphatases to lipid rafts have remained unclear. We show that levels of raft-associated RPTP in the NCAM-deficient brain are reduced, and NCAM redistribution to lipid rafts in response to NCAM activation also induces redistribution of RPTP to lipid rafts via its NCAM association. The combined observations indicate that NCAM plays a role in recruiting NCAM-associated RPTP to lipid rafts via NCAM palmitoylation (Niethammer et al., 2002; Fig. 9) or via NCAM interaction with GPI-anchored components of lipid rafts, such as the GPI-linked GDNF receptor (Paratcha et al., 2003). Investigations on the regulatory mechanisms underlying palmitoylation will be required to understand the subcellular compartment-specific distribution of the NCAM–RPTP–p59fyn complex. Furthermore, the localization of adhesion molecules and receptors will have to be elucidated in view of lipid rafts heterogeneities.
5 e) z5 j; _: S  `. i" Y1 Q
! u+ M* L" c* t' `Potential role of protein tyrosine phosphatases in signaling mediated by other cell adhesion molecules5 A% K5 g# x- @7 G( e7 w4 f; ~+ y+ R

, m$ l6 F3 F. a  \7 hBesides NCAM, activation of L1 and CHL1, other cell adhesion molecules of the immunoglobulin superfamily, also results in the activation of Src family tyrosine kinases (Schmid et al., 2000; Buhusi et al., 2003). As for NCAM, the intracellular domains of L1 and CHL1 do not possess structural motif for protein tyrosine phosphatase activity, suggesting that yet unidentified protein tyrosine phosphatases may be involved. Identification of protein tyrosine phosphatases associated with other cell adhesion molecules of the immunoglobulin superfamily and conjunctions with RPTP-activated integrins (Zeng et al., 2003) will be an important next step in the elucidation of the mechanisms that cell adhesion molecules use differentially to guide cell migration and neurite outgrowth in the developing nervous system.
+ q1 G& s" ^4 T! M$ O/ w( S, j$ T7 a2 L) Z) L- F6 M
Materials and methods6 ?; c- q6 D% w9 F( o" |

; U5 \/ Z& V5 ]. s3 K3 ?/ y+ {/ D9 Z& BAntibodies and toxins
5 U& @4 h; V. z# [4 @
) Z$ I: J6 i5 |/ v; fRabbit polyclonal antibodies against NCAM (Niethammer et al., 2002) were used in immunoprecipitation, immunoblotting, and immunocytochemical experiments; and rat mAbs H28 against mouse NCAM (Gennarini et al., 1984) were used in immunocytochemical experiments. Both antibodies recognize the extracellular domain of all NCAM isoforms. Hybridoma clone H28 was a gift of C. Goridis (Centre National de la Recherche Scientifique UMR 8542, Paris, France). Rabbit antibodies against RPTP were a gift of C.J. Pallen (University of British Columbia, Vancouver, Canada) or were generated as described previously (den Hertog et al., 1994). Rabbit polyclonal antibodies against human erythrocyte spectrin, rabbit polyclonal antibodies against the HA tag, nonspecific rabbit immunoglobulins, and cholera toxin B subunit tagged with fluorescein to label GM1 were obtained from Sigma-Aldrich. Mouse mAbs against the HA tag (clone 12CA5) were obtained from Roche Diagnostics. Rabbit polyclonal antibodies and mouse mAbs against p59fyn protein were purchased from Santa Cruz Biotechnology, Inc. Rabbit polyclonal antibodies against Tyr-527–dephosphorylated or Tyr-416–phosphorylated pp60c-src kinase that cross-react with Tyr-531–dephosphorylated or Tyr-420–phosphorylated p59fyn were obtained from Cell Signaling Technology. Secondary antibodies against rabbit, rat, and mouse Ig coupled to HRP, Cy2, Cy3, or Cy5 were obtained from Dianova.! P% \- H% q1 {5 k( C0 A  Z# W
2 a6 B8 p" S8 C) U
Animals5 y' R0 c# u/ J& X+ b: d* ~, D
) Y2 b( w8 N' s
To compare wild-type and NCAM-deficient mice, C57BL/6J mice and NCAM-deficient mice (Cremer et al., 1994) inbred for at least nine generations onto the C57BL/6J background were used. NCAM-deficient mice were a gift of H. Cremer (Developmental Biology Institute of Marseille, Marseille, France).To compare wild-type and RPTP-deficient mice, RPTP-positive and -negative littermates obtained from heterozygous breeding were used (see online supplemental material).& T( b+ I- b( {

3 R( t5 K" p" N; @( OImage acquisition and manipulation2 {2 y9 d  G2 J  }' p/ U/ i& _2 r
% x; n. U6 ]- C, W) n5 i
Coverslips were embedded in Aqua-Poly/Mount (Polysciences, Inc.). Images were acquired at RT using a confocal laser scanning microscope (model LSM510; Carl Zeiss MicroImaging, Inc.), LSM510 software (version 3; Carl Zeiss MicroImaging, Inc.), and oil Plan-Neofluar 40x objective (NA 1.3; Carl Zeiss MicroImaging, Inc.) at 3x digital zoom. Contrast and brightness of the images were further adjusted in Photo-Paint 9 (Corel Corporation).2 M- S0 I" f5 R5 V
/ \5 c9 z$ m7 q% E; Y" p5 |
Detergent extraction of cultured neurons
1 m  P% B# o. q6 s/ L, g& u, C3 }  q! O8 }" K) g! e5 }
Cells washed in PBS were incubated for 1 min in cold microtubule-stabilizing buffer (2 mM MgCl2, 10 mM EGTA, and 60 mM Pipes, pH 7.0) and extracted 8 min on ice with 1% Triton X-100 in microtubule-stabilizing buffer as described previously (Ledesma et al., 1998). After washing with PBS, cells were fixed with cold 4% formaldehyde in PBS.5 `" b+ ?* s1 r' X- k
; I2 d  Y. h- J' _! [( w; f
Colocalization analysis
7 H# v9 r# D0 m8 w( U% U- C" S  @4 L4 T: h! Z
Colocalization quantification was performed as described previously (Leshchyns'ka et al., 2003). In brief, an NCAM cluster was defined as an accumulation of NCAM labeling with a mean intensity at least 30% higher than background. NCAM clusters were automatically outlined using the threshold function of the Scion Image software (Scion Corporation). Within the outlined areas the mean intensities of NCAM, RPTP, p59fyn, or GM1 labeling associated with NCAM cluster were measured. The same threshold was used for all groups. All experiments were performed two to three times with the same effect. Colocalization profiles were plotted using ImageJ software (National Institutes of Health).9 K. B3 I. J* `: u5 @  x2 ?0 F

3 |; h  r( {. LDNA constructs3 }7 U& W, A9 g8 F3 l) W
/ h1 \7 S$ m. X
Rat NCAM140 and NCAM180/pcDNA3 were a gift of P. Maness (University of North Carolina, Chapel Hill, NC). Rat NCAM120 (a gift of E. Bock, University of Copenhagen, Copenhagen, Denmark) was subcloned into the pcDNA3 vector (Invitrogen) by two EcoRI sites. The EGFP plasmid was purchased from CLONTECH Laboratories, Inc. cDNAs encoding intracellular domains of NCAM140 and NCAM180 were as described previously (Sytnyk et al., 2002; Leshchyns'ka et al., 2003). The plasmid encoding the intracellular domain of RPTP was a gift of C.J. Pallen. Wild-type RPTP, RPTPC433S, and RPTPD2 (containing RPTP residues 1–516 ) were as described previously (den Hertog and Hunter, 1996; Blanchetot and den Hertog, 2000; Buist et al., 2000).
) T6 Q8 K7 J3 q% r+ M% x* {8 ^1 b* }$ G# [  ?* r+ S( H
Online supplemental material
, D1 C8 u0 A( ]. S, M$ `, E0 I: l% X1 c/ k, _4 H
Details on cultures and transfection of hippocampal neurons and CHO cells, immunofluorescence labeling, ELISA and pull-down assay, coimmunoprecipitation, isolation of lipid enriched microdomains, gel electrophoresis, immunoblotting, and generation of RPTP-deficient mice are given in online supplemental material. Online supplemental material is available at http://www.jcb.org/cgi/content/full/jcb.200405073/DC1.
& J: @6 Y, T' Q! T1 u6 j! |
2 r0 [( Q8 H, r0 u0 F3 UAcknowledgments
9 B# m* |8 A4 y, j% |' |9 k& D+ H8 P6 m1 D- d4 `% U
We thank Achim Dahlmann and Eva Kronberg for genotyping and animal care, Drs. Patricia Maness and Elisabeth Bock for NCAM cDNAs, Dr. Catherine J. Pallen for antibodies against RPTP and cDNA encoding the RPTP intracellular domain, Dr. Harold Cremer for NCAM-deficient mice, and Dr. Christo Goridis for hybridoma clone H28.
8 `9 O! U. Q$ g, D3 i; p4 i* ^; L& Y/ {) t  |# a6 G
This work was supported by Zonta Club, Hamburg-Alster (I. Leshchyns'ka), and Deutsche Forschungsgemeinschaft (I. Leshchyns'ka, V. Sytnyk, and M. Schachner).
% ?" t+ R6 m0 G, I& A* v+ ]4 b
/ ^" Q3 T. L9 e! R7 F( a8 D1 y6 uSubmitted: 12 May 2004
! A( `# C+ |9 j1 w9 i! P
% Q; \  y) w- V8 f2 j+ a$ ?Accepted: 11 November 2004
9 Q) h. k+ @" I+ p
6 S( M' ?+ f( zReferences
5 n3 t: }) ~4 T1 O! @" t; x  r- j7 k
2 Z* X* U# a! I4 H) p4 u1 wBeggs, H.E., P. Soriano, and P.F. Maness. 1994. NCAM-dependent neurite outgrowth is inhibited in neurons from Fyn-minus mice. J. Cell Biol. 127:825–833.
1 Q/ p( T2 C- |# u$ Q9 V5 t3 @  |# Y. e8 z. A% w8 ?
Beggs, H.E., S.C. Baragona, J.J. Hemperly, and P.F. Maness. 1997. NCAM140 interacts with the focal adhesion kinase p125(fak) and the SRC-related tyrosine kinase p59(fyn). J. Biol. Chem. 272:8310–8319.# ~/ h# S5 c- [6 k& U5 J
- K7 [; ?3 D# z* s- J% j
Bhandari, V., K.L. Lim, and C.J. Pallen. 1998. Physical and functional interactions between receptor-like protein-tyrosine phosphatase alpha and p59fyn. J. Biol. Chem. 273:8691–8698.$ l/ ?8 [0 b1 o: @5 a- f% I& l. ?% Q

/ c' z3 K: c& l( C5 L+ ^Blanchetot, C., and J. den Hertog. 2000. Multiple interactions between receptor protein-tyrosine phosphatase (RPTP) alpha and membrane-distal protein-tyrosine phosphatase domains of various RPTPs. J. Biol. Chem. 275:12446–12452.
7 z3 u# f6 k/ b/ ~" d
/ t! Q$ m/ o0 x# ^Brown, M.T., and J.A. Cooper. 1996. Regulation, substrates and functions of src. Biochim. Biophys. Acta. 1287:121–149.
- D, v. W+ L% j5 Q2 k) F2 |. d/ V8 w7 ?4 a9 B
Buhusi, M., B.R. Midkiff, A.M. Gates, M. Richter, M. Schachner, and P.F. Maness. 2003. Close homolog of L1 is an enhancer of integrin-mediated cell migration. J. Biol. Chem. 278:25024–25031.
6 {- R# i/ W& q  U
+ V9 r; e  e0 i" v, E  o- c7 pBuist, A., C. Blanchetot, L.G. Tertoolen, and J. den Hertog. 2000. Identification of p130cas as an in vivo substrate of receptor protein-tyrosine phosphatase alpha. J. Biol. Chem. 275:20754–20761.
1 w- M0 a7 X7 J, R! X4 O; b$ l- U# ?/ M# T/ p2 G" ?- g' J( m2 D
Cavallaro, U., J. Niedermeyer, M. Fuxa, and G. Christofori. 2001. N-CAM modulates tumour-cell adhesion to matrix by inducing FGF-receptor signalling. Nat. Cell Biol. 3:650–657.- B  s0 Q0 }# s7 [6 q8 F6 }

# Z6 n, \. R0 X& K5 |0 b$ ACremer, H., R. Lange, A. Christoph, M. Plomann, G. Vopper, J. Roes, R. Brown, S. Baldwin, P. Kraemer, S. Scheff, et al. 1994. Inactivation of the N-CAM gene in mice results in size reduction of the olfactory bulb and deficits in spatial learning. Nature. 367:455–459.
. H; W3 j( b: d* \9 v% u, Z  I3 D. ]6 A4 f" W4 ]7 H& D
Crossin, K.L., and L.A. Krushel. 2000. Cellular signaling by neural cell adhesion molecules of the immunoglobulin superfamily. Dev. Dyn. 218:260–279.  u# T0 m4 S' e4 r

% ~! D9 F* R3 c  [6 E3 TDe Matteis, M.A., and J.S. Morrow. 2000. Spectrin tethers and mesh in the biosynthetic pathway. J. Cell Sci. 113:2331–2343.
6 Y& N1 a! ]3 N; q4 t4 H+ M( B$ C
den Hertog, J., and T. Hunter. 1996. Tight association of GRB2 with receptor protein-tyrosine phosphatase alpha is mediated by the SH2 and C-terminal SH3 domains. EMBO J. 15:3016–3027.' W( ]; A: f9 c

1 Q0 w, c1 r3 g9 J& b+ A: _3 @den Hertog, J., C.E. Pals, M.P. Peppelenbosch, L.G. Tertoolen, S.W. de Laat, and W. Kruijer. 1993. Receptor protein tyrosine phosphatase alpha activates pp60c-src and is involved in neuronal differentiation. EMBO J. 12:3789–3798.- \& v6 k; ^( Z: C
' i( I9 X  ]3 |$ R1 @, |
den Hertog, J., S. Tracy, and T. Hunter. 1994. Phosphorylation of receptor protein-tyrosine phosphatase alpha on Tyr789, a binding site for the SH3-SH2-SH3 adaptor protein GRB-2 in vivo. EMBO J. 13:3020–3032.
. T4 d0 v' j* ?' m! _+ \7 v$ `, @" |$ s
den Hertog, J., J. Sap, C.E. Pals, J. Schlessinger, and W. Kruijer. 1995. Stimulation of receptor protein-tyrosine phosphatase alpha activity and phosphorylation by phorbol ester. Cell Growth Differ. 6:303–307.$ q% F( v; m1 l" y9 _% D

4 ~2 j' R7 g( I8 |0 Q9 N1 HFilipp, D., J. Zhang, B.L. Leung, A. Shaw, S.D. Levin, A. Veillette, and M. Julius. 2003. Regulation of Fyn through translocation of activated Lck into lipid rafts. J. Exp. Med. 197:1221–1227.- h$ F6 G. f6 O1 ?% ^3 `
) {* r$ V6 H5 a8 Z( U
Gennarini, G., M. Hirn, H. Deagostini-Bazin, and C. Goridis. 1984. Studies on the transmembrane disposition of the neural cell adhesion molecule N-CAM. The use of liposome-inserted radioiodinated N-CAM to study its transbilayer orientation. Eur. J. Biochem. 142:65–73.% M+ @9 B1 {: U1 z3 f/ A7 x
( c! n- t* Z7 F
Harder, K.W., N.P. Moller, J.W. Peacock, and F.R. Jirik. 1998. Protein-tyrosine phosphatase alpha regulates Src family kinases and alters cell-substratum adhesion. J. Biol. Chem. 273:31890–31900.2 u9 n0 S0 ]* S- j: q

' f- x! H. l& Z- [3 m. I" j) }- THe, Q., and K.F. Meiri. 2002. Isolation and characterization of detergent-resistant microdomains responsive to NCAM-mediated signaling from growth cones. Mol. Cell. Neurosci. 19:18–31.
' G7 o  L, R+ ~. j
! ]. G0 z! W9 w8 ?/ t, `) xHelmke, S., K. Lohse, K. Mikule, M.R. Wood, and K.H. Pfenninger. 1998. SRC binding to the cytoskeleton, triggered by growth cone attachment to laminin, is protein tyrosine phosphatase-dependent. J. Cell Sci. 111:2465–2475.8 y- W4 Y, f$ K6 ?% u

$ M; O! W: f9 T: q' @Hubbard, S.R. 1999. Src autoinhibition: let us count the ways. Nat. Struct. Biol. 6:711–714.
3 D5 v- V" O1 U6 I5 a  w1 J6 Q: M, {" Z0 k
Iida, N., V.B. Lokeshwar, and L.Y. Bourguignon. 1994. Mapping the fodrin binding domain in CD45, a leukocyte membrane-associated tyrosine phosphatase. J. Biol. Chem. 269:28576–28583.
& T6 \2 h& a, {/ [) U4 T3 Z- i
: ^* p9 F5 G# g) p4 V& `Juliano, R.L. 2002. Signal transduction by cell adhesion receptors and the cytoskeleton: functions of integrins, cadherins, selectins, and immunoglobulin-superfamily members. Annu. Rev. Pharmacol. Toxicol. 42:283–323.
( |% g3 N2 u8 r% ~- q% l; W" O7 p( [2 W" ^9 S' I4 W9 |
Kamiguchi, H., and V. Lemmon. 2000. IgCAMs: bidirectional signals underlying neurite growth. Curr. Opin. Cell Biol. 12:598–605.! Q/ ?8 r- o5 h3 `

  C/ n2 O6 U( U! M. k6 B, uKaplan, R., B. Morse, K. Huebner, C. Croce, R. Howk, M. Ravera, G. Ricca, M. Jaye, and J. Schlessinger. 1990. Cloning of three human tyrosine phosphatases reveals a multigene family of receptor-linked protein-tyrosine-phosphatases expressed in brain. Proc. Natl. Acad. Sci. USA. 87:7000–7004.
4 U3 s. @- `( `6 Y
" i8 F  a7 R" ~! {Kolkova, K., V. Novitskaya, N. Pedersen, V. Berezin, and E. Bock. 2000. Neural cell adhesion molecule-stimulated neurite outgrowth depends on activation of protein kinase C and the Ras-mitogen-activated protein kinase pathway. J. Neurosci. 20:2238–2246.
1 s" c1 `8 ?; u( |1 M# ^' h2 w' \5 n
Kramer, E.M., C. Klein, T. Koch, M. Boytinck, and J. Trotter. 1999. Compartmentation of Fyn kinase with glycosylphosphatidylinositol-anchored molecules in oligodendrocytes facilitates kinase activation during myelination. J. Biol. Chem. 274:29042–29049.0 N. p% F) H" k3 g- H% @. G5 c0 s

$ B& M; N& q8 C0 h9 JKrueger, N.X., M. Streuli, and H. Saito. 1990. Structural diversity and evolution of human receptor-like protein tyrosine phosphatases. EMBO J. 9:3241–3252.
( X3 I+ L7 L/ U& A# x/ c! f* k" b4 q, E4 i+ x+ w
Ledesma, M.D., K. Simons, and C.G. Dotti. 1998. Neuronal polarity: essential role of protein-lipid complexes in axonal sorting. Proc. Natl. Acad. Sci. USA. 95:3966–3971.. {, ~, S  v8 `6 B2 W& m: @

$ ]" W4 B. v8 b/ o2 ]' d! x7 }Leshchyns'ka, I., V. Sytnyk, J.S. Morrow, and M. Schachner. 2003. Neural cell adhesion molecule (NCAM) association with PKC?2 via ?I spectrin is implicated in NCAM-mediated neurite outgrowth. J. Cell Biol. 161:625–639.
3 Z8 ^* d* o% F/ |: R7 m7 S& Q  e. S- w! f7 k
Lokeshwar, V.B., and L.Y. Bourguignon. 1992. Tyrosine phosphatase activity of lymphoma CD45 (GP180) is regulated by a direct interaction with the cytoskeleton. J. Biol. Chem. 267:21551–21557.
% ^) n# t. g( ~" o! l" j+ F) x
! P$ a. S; K  v) B) T. m; GNiethammer, P., M. Delling, V. Sytnyk, A. Dityatev, K. Fukami, and M. Schachner. 2002. Cosignaling of NCAM via lipid rafts and the FGF receptor is required for neuritogenesis. J. Cell Biol. 157:521–532.- B0 r7 A0 b3 W. c! N) h1 m
2 q+ A# z* H% G% }: y9 U  d
Paratcha, G., F. Ledda, and C.F. Ibanez. 2003. The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands. Cell. 113:867–879.0 v" C; q* k/ }$ F: n

8 t; m1 M% W  XPetrone, A., and J. Sap. 2000. Emerging issues in receptor protein tyrosine phosphatase function: lifting fog or simply shifting? J. Cell Sci. 113:2345–2354.( U6 G$ _1 F: U1 H3 X

5 n% L- u1 y, _9 z! mPetrone, A., F. Battaglia, C. Wang, A. Dusa, J. Su, D. Zagzag, R. Bianchi, P. Casaccia-Bonnefil, O. Arancio, and J. Sap. 2003. Receptor protein tyrosine phosphatase alpha is essential for hippocampal neuronal migration and long-term potentiation. EMBO J. 22:4121–4131.2 |- T# q1 M! w7 G
& I$ ~( c+ Z3 Q4 n
Ponniah, S., D.Z. Wang, K.L. Lim, and C.J. Pallen. 1999. Targeted disruption of the tyrosine phosphatase PTPalpha leads to constitutive downregulation of the kinases Src and Fyn. Curr. Biol. 9:535–538.
9 `/ N( c. P. x2 a$ R
% S1 n; i6 J( _- g* k/ ~' MSchmid, R.S., R.D. Graff, M.D. Schaller, S. Chen, M. Schachner, J.J. Hemperly, and P.F. Maness. 1999. NCAM stimulates the Ras-MAPK pathway and CREB phosphorylation in neuronal cells. J. Neurobiol. 38:542–558.
1 Z! |: r6 J6 I) S! r+ L# g9 ~# f1 Q* ?; m7 [0 ?) R
Schmid, R.S., W.M. Pruitt, and P.F. Maness. 2000. A MAP kinase-signaling pathway mediates neurite outgrowth on L1 and requires Src-dependent endocytosis. J. Neurosci. 20:4177–4188.
1 |: U0 m7 N+ Q- a" f
1 ]2 b2 ^7 ^2 H) f& g" KSu, J., L.T. Yang, and J. Sap. 1996. Association between receptor protein-tyrosine phosphatase RPTPalpha and the Grb2 adaptor. Dual Src homology (SH) 2/SH3 domain requirement and functional consequences. J. Biol. Chem. 271:28086–28096.5 ^: `% A7 }6 G# ~0 @
0 U' x" N  |! D6 c! u
Sytnyk, V., I. Leshchyns'ka, M. Delling, G. Dityateva, A. Dityatev, and M. Schachner. 2002. Neural cell adhesion molecule promotes accumulation of TGN organelles at sites of neuron-to-neuron contacts. J. Cell Biol. 159:649–661.
2 ]3 U. A* \# A% S% t1 g8 K$ W6 s6 J( x; r
Thomas, S.M., and J.S. Brugge. 1997. Cellular functions regulated by Src family kinases. Annu. Rev. Cell Dev. Biol. 13:513–609.$ V; _7 p- X; s" t% U2 f  {
+ ^3 r' c- @/ R/ B! u0 x1 N
Tracy, S., P. van der Geer, and T. Hunter. 1995. The receptor-like protein-tyrosine phosphatase, RPTP alpha, is phosphorylated by protein kinase C on two serines close to the inner face of the plasma membrane. J. Biol. Chem. 270:10587–10594.% M- v, Y; Y% F! `( \" K" O# N' ?

  B) Z2 a% s7 l8 }5 x6 Dvan't Hof, W., and M.D. Resh. 1997. Rapid plasma membrane anchoring of newly synthesized p59fyn: selective requirement for NH2-terminal myristoylation and palmitoylation at cysteine-3. J. Cell Biol. 136:1023–1035.
6 K7 ?1 Z8 E$ }! w9 w
2 y' @$ M, `3 h1 U: S0 F. ivon Wichert, G., G. Jiang, A. Kostic, K. De Vos, J. Sap, and M.P. Sheetz. 2003. RPTP- acts as a transducer of mechanical force on v/?3-integrin–cytoskeleton linkages. J. Cell Biol. 161:143–153.
) g* M# I% M& y" L( _2 z( a/ }3 H/ ^4 L
Walsh, F.S., and P. Doherty. 1997. Neural cell adhesion molecules of the immunoglobulin superfamily: role in axon growth and guidance. Annu. Rev. Cell Dev. Biol. 13:425–456." ]4 e9 ?2 D2 y; Q9 M) j) v

3 J) t- @2 B. g: C- g- m: ^Wang, Y., and C.J. Pallen. 1991. The receptor-like protein tyrosine phosphatase HPTP alpha has two active catalytic domains with distinct substrate specificities. EMBO J. 10:3231–3237.) Z! N. d/ l( \& J% N. U6 q; E

( C! b$ t8 a, l' VWilliams, E.J., P. Doherty, G. Turner, R.A. Reid, J.J. Hemperly, and F.S. Walsh. 1992. Calcium influx into neurons can solely account for cell contact–dependent neurite outgrowth stimulated by transfected L1. J. Cell Biol. 119:883–892.
4 m- Q2 h1 u0 H% P2 j2 E2 x- P" D7 W+ F( B% p# Y
Wu, L., A. Buist, J. den Hertog, and Z.Y. Zhang. 1997. Comparative kinetic analysis and substrate specificity of the tandem catalytic domains of the receptor-like protein-tyrosine phosphatase alpha. J. Biol. Chem. 272:6994–7002.
# Q: a# {' i0 Q- d/ s3 a9 q9 a: y7 p" X. @) x+ i) ]" d
Yang, L.T., K. Alexandropoulos, and J. Sap. 2002. c-SRC mediates neurite outgrowth through recruitment of Crk to the scaffolding protein Sin/Efs without altering the kinetics of ERK activation. J. Biol. Chem. 277:17406–17414." |5 t. F7 L$ L! L/ }
: \; T* v, U% l: n+ n* h3 o( M
Zeng, L., L. D'Alessandri, M.B. Kalousek, L. Vaughan, and C.J. Pallen. 1999. Protein tyrosine phosphatase  (PTP) and contactin form a novel neuronal receptor complex linked to the intracellular tyrosine kinase fyn. J. Cell Biol. 147:707–714.
. j4 r9 e5 }" c# A" v% O% H  B7 M* N, [  i- `
Zeng, L., X. Si, W.P. Yu, H.T. Le, K.P. Ng, R.M. Teng, K. Ryan, D.Z. Wang, S. Ponniah, and C.J. Pallen. 2003. PTP regulates integrin-stimulated FAK autophosphorylation and cytoskeletal rearrangement in cell spreading and migration. J. Cell Biol. 160:137–146.
" k8 [( z" @' F0 ?# k- B9 D3 L+ v/ U1 m: s) y* d3 ]
Zheng, X.M., Y. Wang, and C.J. Pallen. 1992. Cell transformation and activation of pp60c-src by overexpression of a protein tyrosine phosphatase. Nature. 359:336–339.
, ~  w6 i( U8 z' j3 T* Q
" ^# L$ V5 v- N$ fZheng, X.M., R.J. Resnick, and D. Shalloway. 2000. A phosphotyrosine displacement mechanism for activation of Src by PTPalpha. EMBO J. 19:964–978.: E, P- ^3 j$ o  w4 V9 i& _( m

( H$ v" z& E$ CZheng, X.M., R.J. Resnick, and D. Shalloway. 2002. Mitotic activation of protein-tyrosine phosphatase alpha and regulation of its Src-mediated transforming activity by its sites of protein kinase C phosphorylation. J. Biol. Chem. 277:21922–21929.
1 l$ t) i* x# L5 Y/ \8 d+ ?, R$ w+ E0 U( P- u
This article has been cited by other articles: (Search Google Scholar for Other Citing Articles)
( I, H' D% ]1 n& w, v: i4 e5 ^2 S" b! y# {5 `" y5 Q6 E# h
Tiran, Z., Peretz, A., Sines, T., Shinder, V., Sap, J., Attali, B., Elson, A. (2006). Tyrosine Phosphatases {varepsilon} and {alpha} Perform Specific and Overlapping Functions in Regulation of Voltage-gated Potassium Channels in Schwann Cells. Mol. Biol. Cell 17: 4330-43425 l, v, m! a1 z' _
' ]3 `4 D2 ~$ u# f( o* Y
Sytnyk, V., Leshchyns'ka, I., Nikonenko, A. G., Schachner, M. (2006). NCAM promotes assembly and activity-dependent remodeling of the postsynaptic signaling complex. J. Cell Biol. 174: 1071-1085" j3 h' Q- s5 x8 l& e& j/ D% w; ^
$ \9 A, Y3 h$ ?
Santuccione, A., Sytnyk, V., Leshchyns'ka, I., Schachner, M. (2005). Prion protein recruits its neuronal receptor NCAM to lipid rafts to activate p59fyn and to enhance neurite outgrowth. J. Cell Biol. 169: 341-354(Vsevolod Bodrikov1, Iryna Leshchyns'ka1,)

Rank: 2

积分
69 
威望
69  
包包
1788  
沙发
发表于 2015-5-31 12:42 |只看该作者
不对,就是碗是铁的,里边没饭你吃啥去?  

Rank: 2

积分
72 
威望
72  
包包
1859  
藤椅
发表于 2015-6-2 09:10 |只看该作者
昨晚多几分钟的准备,今天少几小时的麻烦。  

Rank: 2

积分
77 
威望
77  
包包
1964  
板凳
发表于 2015-6-9 07:14 |只看该作者
干细胞之家微信公众号
干细胞分化技术

Rank: 2

积分
107 
威望
107  
包包
1889  
报纸
发表于 2015-6-23 19:20 |只看该作者
我帮你 喝喝  

Rank: 2

积分
75 
威望
75  
包包
2193  
地板
发表于 2015-7-14 18:31 |只看该作者
做一个,做好了,请看  

Rank: 2

积分
69 
威望
69  
包包
1788  
7
发表于 2015-7-18 17:24 |只看该作者
干细胞从业人员  

Rank: 2

积分
64 
威望
64  
包包
1734  
8
发表于 2015-8-5 14:43 |只看该作者
我在努力中  

Rank: 2

积分
89 
威望
89  
包包
1794  
9
发表于 2015-8-18 08:01 |只看该作者
偶真幸运哦...  

Rank: 2

积分
104 
威望
104  
包包
1772  
10
发表于 2015-8-21 21:36 |只看该作者
也许似乎大概是,然而未必不见得。  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-4-26 16:31

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.