j. biol. chem.-1997-morley-17887-93

8
Involvement of Stress-activated Protein Kinase and p38/RK Mitogen-activated Protein Kinase Signaling Pathways in the Enhanced Phosphorylation of Initiation Factor 4E in NIH 3T3 Cells* (Received for publication, January 27, 1997, and in revised form, April 7, 1997) Simon J. Morley‡ and Linda McKendrick  From the Biochemistry Laboratory, School of Biological Sciences, University of Sussex, Falmer,  Brighton BN1 9QG, United Kingdom The initiation factor (eIF) 4E is regulated by modulat- ing both the phosphorylation and the availability of the protein to participate in the initiation process. Here we show that either serum treatment or activation of the stres s-act ivate d prote in kinase (JNK/ SAPK) led to en- hanced phosphorylation of eIF4E in quiescent NIH 3T3 cell s. Although the immunosu ppre ssant, rapa myci n, was found to stabilize the association of eIF4E with its negat ive regulato r, 4E-BP1, this drug did not prev ent the early effects of serum stimulation on the overall rate of translation, polysome formation, the phosphorylation status of eIF4E, or the recruitment of eIF4E into the eIF4F comple x. Howeve r, the rapid enh anc ement of eIF4E pho sph ory lat ion in res pon se to ser um was lar gel y prevented by the inhibitor of mitogen-activated protein (MAP) kin ase act iva tio n, PD9 805 9. Act iva tio n of the JNK/SAPK signaling pathway with anisomycin resulted in enhanced phosphorylation of eIF4E, which was pre- vented by either rapamycin or the highly specific p38 MAP kinase inhibitor, SB203580. These data illustrate that multi ple sig nal ing pat hwa ys, inc lud ing tho se of dis - tinct members of the MAP kina se fami ly, mediat e the phosp horyl ation of eIF4E and that the associat ion of eIF4E with 4E-BP1 does not necessarily prevent phos- phorylation of eIF4E in vivo. Control of polypeptide synthesis plays an important role in cell proliferation, with physiological regulation of protein syn- thesis almost always exerted at the level of polypeptide chain initiation (reviewed in Refs. 1 and 2). This phase is regulated, in part, by the phosphorylation of initiation factors involved in binding mRNA to the 40 S ribosomal subunit, a step which appears to be rate-limiting in many cell systems (1–6). The cap structure present at the 5 end of mRNA facilitates its binding to the ribosome, a process mediated by at least three initiation factors (eIF4A, 1 -4B, and -4F) and ATP hydrolysis (1–4, 7, 8). eIF4F is a cap-binding protein complex composed of three sub- units; eIF4E, which specifically recognizes the cap structure (9); eIF4A, an ATP-dependent, single strand RNA-binding pro- tein with helicase activity (4, 8); and eIF4G, which acts as a bridging molecule between eIF4E and the 40 S ribosome, prob- ably via eIF3 (10, 11). It is believed that eIF4F functions to unwind secondary structure in the mRNA 5-untranslated re- gion to facilitate binding to the 40 S ribosomal subunit (1–6). Consistent with its proposed regulatory role, eIF4E exists in both phosphorylated and non-phosphorylated forms. Although increased levels of eIF4E phosphorylation have been directly correlated with enhanced rates of translation in a variety of cell types (reviewed in Ref. 1), it is still not clear how phosphoryl- ation of eIF4E modulates its activity. While there may be a direct effect of phosphorylation on cap structure recognition in vitro (12), phosphorylation of eIF4E  in vivo  can also be corre- lated with enhanced interaction with other components of the eIF4F complex (13, 14). Two additional proteins (4E-BP1 and 4E-BP2), which inter- act with eIF4E and inhibit cap structure-dependent transla- tion, have been identified as downstream signaling targets (15, 16). Phosphorylation of 4E-BP1 disrupts its interaction with eIF4E, liberating eIF4E to interact with a conserved hydropho- bic region of eIF4G. A similar sequence found in 4E-BP1 is inv olved in bin din g to eIF4E and compe tes with eIF4G for eIF4E binding (17). It is believed that the phosphorylation of 4E-BP 1 and conse quent liber ation of eIF4 E lead to the up- regulation of translation (1, 15). It has also been claimed that assoc iat ion wit h 4E- BP1 pre vents the pho sph ory lation of eIF4E by protein kinase C  in vitro  (18). In several cell types, the phosphorylation of 4E-BP1 is inhibited by the immunosup- pre ssa nt, rap amy cin , whi ch pre vents the act iva tion of the p70 S6K signa ling path way and stabiliz es the intera ction be- tween eIF4E and 4E-BP1 (see Refs. 1, 4, and 16, and references therein). However, rapamycin does not prevent the phospho- rylation of eIF4E in primary pig T cells (19),  Xenopus  oocytes (20), CHO.T cells in response to insulin (21), or NIH 3T3 cells in response to serum (22). We have examined the signal transduction pathways that are involved in the enhanced phosphorylation of eIF4E and its recruitment to ribosomes. Our data indicate that, in NIH 3T3 cells, eIF4E phosphorylation is enhanced in response either to serum or to activation of the JNK/SAPK and p38/RK signaling pathways with anisomycin. In response to serum stimulation, eIF4 E phos phory latio n is large ly medi ated via the class ical MAP kinase pathway, and is independent of both p70 S6K sig- naling and association of eIF4E with 4E-BP1. However, follow- ing anisomycin treatment, phosphorylation of eIF4E via JNK/ SAPK is depe ndent upon p70 S6K and the p38/ RK signa ling path ways . Thes e data suggest that the phos phory lati on of eIF4E and 4E-BP1 can be regulated independently and that each may play a direct role in regulating translational initia- tion in vivo. * This work was supported in part by Grant 045619/Z/95 from The Wellcome Trust. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement ” in accordance with 18 U.S.C. Section 1734 solely to indic ate this fact. ‡ Se nior Research Fello w of The Well come Trust. To whom corre- spond ence should be addressed. Tel.: 44-127 3-6785 44; Fax: 44-1273- 678433; E-mail: [email protected]. 1 The abbreviat ion s use d are: eIF, euk aryoti c ini tiatio n fac tor ; m 7 GTP, 7-methyl guanosine triphosphate; PMA, phorbol 12-myristate 13-acetate; Mops, 3-(  N -morpholino)propanesulfonic acid; PAGE, poly- acrylamide gel electrophoresis; VSIEF, vertical-slab isoelectric focus- ing; GST, glutathione-  S-transferase; JNK/SAPK, c-Jun NH 2 -terminal kinase/stress-activated protein kinase; FCS, fetal calf serum. THE JOURNAL OF BIOLOGICAL CHEMISTRY  Vol. 272, No. 28, Issue of July 11, pp. 17887–17893, 1997  © 1997 by The American Society for Biochemistry and Molecular Biology, Inc.  Printed in U.S.A. This paper is available on line at http://www.jbc.org  17887   b  y  g  u  e  s  t   o F  e  b r  u  a r  y  6  , 2  0 1  5 h  t   t   p  :  /   /   w  w  w  .  j   b  c  .  o r  g  /  D  o  w n l   o  a  d  e  d f  r  o m  

Upload: amandbhaskar

Post on 08-Mar-2016

214 views

Category:

Documents


0 download

DESCRIPTION

Involvement of Stress-activated Protein Kinase and p38/RKMitogen-activated Protein Kinase Signaling Pathways in theEnhanced Phosphorylation of Initiation Factor 4E in NIH 3T3 Cells

TRANSCRIPT

Page 1: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 1/8

Involvement of Stress-activated Protein Kinase and p38/RK Mitogen-activated Protein Kinase Signaling Pathways in theEnhanced Phosphorylation of Initiation Factor 4E in NIH 3T3 Cells*

(Received for publication, January 27, 1997, and in revised form, April 7, 1997)

Simon J. Morley‡ and Linda McKendrick From the Biochemistry Laboratory, School of Biological Sciences, University of Sussex, Falmer, Brighton BN1 9QG, United Kingdom

The initiation factor (eIF) 4E is regulated by modulat-ing both the phosphorylation and the availability of theprotein to participate in the initiation process. Here weshow that either serum treatment or activation of thestress-activated protein kinase (JNK/SAPK) led to en-hanced phosphorylation of eIF4E in quiescent NIH 3T3cells. Although the immunosuppressant, rapamycin,was found to stabilize the association of eIF4E with itsnegative regulator, 4E-BP1, this drug did not preventthe early effects of serum stimulation on the overall rateof translation, polysome formation, the phosphorylationstatus of eIF4E, or the recruitment of eIF4E into theeIF4F complex. However, the rapid enhancement of eIF4E phosphorylation in response to serum was largelyprevented by the inhibitor of mitogen-activated protein(MAP) kinase activation, PD98059. Activation of theJNK/SAPK signaling pathway with anisomycin resultedin enhanced phosphorylation of eIF4E, which was pre-vented by either rapamycin or the highly specific p38MAP kinase inhibitor, SB203580. These data illustratethat multiple signaling pathways, including those of dis-tinct members of the MAP kinase family, mediate thephosphorylation of eIF4E and that the association of eIF4E with 4E-BP1 does not necessarily prevent phos-phorylation of eIF4E   in vivo.

Control of polypeptide synthesis plays an important role in

cell proliferation, with physiological regulation of protein syn-

thesis almost always exerted at the level of polypeptide chain

initiation (reviewed in Refs. 1 and 2). This phase is regulated,

in part, by the phosphorylation of initiation factors involved in

binding mRNA to the 40 S ribosomal subunit, a step which

appears to be rate-limiting in many cell systems (1–6). The cap

structure present at the 5 end of mRNA facilitates its binding 

to the ribosome, a process mediated by at least three initiation

factors (eIF4A,1 -4B, and -4F) and ATP hydrolysis (1–4, 7, 8).

eIF4F is a cap-binding protein complex composed of three sub-

units; eIF4E, which specifically recognizes the cap structure

(9); eIF4A, an ATP-dependent, single strand RNA-binding pro-

tein with helicase activity (4, 8); and eIF4G, which acts as a

bridging molecule between eIF4E and the 40 S ribosome, prob-

ably via eIF3 (10, 11). It is believed that eIF4F functions to

unwind secondary structure in the mRNA 5-untranslated re-

gion to facilitate binding to the 40 S ribosomal subunit (1–6).

Consistent with its proposed regulatory role, eIF4E exists in

both phosphorylated and non-phosphorylated forms. Although

increased levels of eIF4E phosphorylation have been directly

correlated with enhanced rates of translation in a variety of celltypes (reviewed in Ref. 1), it is still not clear how phosphoryl-

ation of eIF4E modulates its activity. While there may be a

direct effect of phosphorylation on cap structure recognition in

vitro   (12), phosphorylation of eIF4E  in vivo  can also be corre-

lated with enhanced interaction with other components of the

eIF4F complex (13, 14).

Two additional proteins (4E-BP1 and 4E-BP2), which inter-

act with eIF4E and inhibit cap structure-dependent transla-

tion, have been identified as downstream signaling targets (15,

16). Phosphorylation of 4E-BP1 disrupts its interaction with

eIF4E, liberating eIF4E to interact with a conserved hydropho-

bic region of eIF4G. A similar sequence found in 4E-BP1 is

involved in binding to eIF4E and competes with eIF4G for

eIF4E binding (17). It is believed that the phosphorylation of 4E-BP1 and consequent liberation of eIF4E lead to the up-

regulation of translation (1, 15). It has also been claimed that

association with 4E-BP1 prevents the phosphorylation of 

eIF4E by protein kinase C  in vitro  (18). In several cell types,

the phosphorylation of 4E-BP1 is inhibited by the immunosup-

pressant, rapamycin, which prevents the activation of the

p70S6K  signaling pathway and stabilizes the interaction be-

tween eIF4E and 4E-BP1 (see Refs. 1, 4, and 16, and references

therein). However, rapamycin does not prevent the phospho-

rylation of eIF4E in primary pig T cells (19),  Xenopus  oocytes

(20), CHO.T cells in response to insulin (21), or NIH 3T3 cells

in response to serum (22).

We have examined the signal transduction pathways that

are involved in the enhanced phosphorylation of eIF4E and its

recruitment to ribosomes. Our data indicate that, in NIH 3T3

cells, eIF4E phosphorylation is enhanced in response either to

serum or to activation of the JNK/SAPK and p38/RK signaling 

pathways with anisomycin. In response to serum stimulation,

eIF4E phosphorylation is largely mediated via the classical

MAP kinase pathway, and is independent of both p70S6K  sig-

naling and association of eIF4E with 4E-BP1. However, follow-

ing anisomycin treatment, phosphorylation of eIF4E via JNK/ 

SAPK is dependent upon p70S6K  and the p38/RK signaling 

pathways. These data suggest that the phosphorylation of 

eIF4E and 4E-BP1 can be regulated independently and that

each may play a direct role in regulating translational initia-

tion in vivo.

* This work was supported in part by Grant 045619/Z/95 from TheWellcome Trust. The costs of publication of this article were defrayed inpart by the payment of page charges. This article must therefore behereby marked “advertisement” in accordance with 18 U.S.C. Section1734 solely to indicate this fact.

‡ Senior Research Fellow of The Wellcome Trust. To whom corre-spondence should be addressed. Tel.: 44-1273-678544; Fax: 44-1273-678433; E-mail: [email protected].

1 The abbreviations used are: eIF, eukaryotic initiation factor;m7GTP, 7-methyl guanosine triphosphate; PMA, phorbol 12-myristate13-acetate; Mops, 3-( N -morpholino)propanesulfonic acid; PAGE, poly-acrylamide gel electrophoresis; VSIEF, vertical-slab isoelectric focus-ing; GST, glutathione- S-transferase; JNK/SAPK, c-Jun NH2-terminalkinase/stress-activated protein kinase; FCS, fetal calf serum.

THE  JOURNAL OF  BIOLOGICAL  CHEMISTRY    Vol. 272, No. 28, Issue of July 11, pp. 17887–17893, 1997 © 1997 by The American Society for Biochemistry and Molecular Biology, Inc.   Printed in U.S.A.

This paper is available on line at http://www.jbc.org   17887

Page 2: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 2/8

EXPERIMENTAL PROCEDURES

Chemicals and Biochemicals—Materials for tissue culture were fromLife Technologies, Inc. [35S] methionine was from ICN, Immobilonpolyvinylidine difluoride was from Millipore, and m7GTP-Sepharose

was from Pharmacia Biotech Inc. Microcystin was from Calbiochem;unless otherwise stated, all other chemicals were from Sigma. Anti-

serum to 4E-BP1 (PHAS-I) was kindly provided by Dr. J. Lawrence, Jr.(Washington University School of Medicine, St. Louis, MO) and Prof. R.Denton (Department of Biochemistry, Bristol, UK) and the plasmid

encoding the glutathione- S-transferase fusion protein GST-c-Jun-(1–

79) was provided by Dr. M. Karin (University of California at SanDiego, San Diego, CA). Rapamycin was a kind gift from Dr. J. Kay

(Department of Biochemistry, Sussex, UK), PD098059 was from Parke-Davis, RO31-8220 was from the Roche Research Center, UK, and

SB203580 was a gift from SmithKline Beecham (King of Prussia, PA).Tissue Culture—NIH 3T3 cells were grown in Dulbecco’s modified

Eagle’s medium with Glutamax™ supplemented with 10% fetal calf 

serum (FCS). Prior to treatment with rapamycin, RO31-8220, PD98059,or SB203580, cells were grown to 80% confluence and then serum-starved in 0.5% FCS for 48 h. Treatments with agonists/inhibitors were

as described in individual figure legends. Cells were then washed andextracts prepared as described below.

 Preparation of Cell Extracts—Following treatment, the medium was

removed and plates of cells were transferred to ice. Cells were scrapedinto 0.5 ml of Buffer A (50 mM Mops-KOH, pH 7.4, 2.5 mM EGTA, 1 mM

EDTA, 40 mM  -glycerophosphate, 1  M   microcystin, 120 mM  NaCl, 7mM 2-mercaptoethanol, 2 mM benzamidine, 1 mM phenylmethylsulfonyl

fluoride, 0.1 mM  GTP, 2 mM  Na3 VO4), isolated by centrifugation, andwashed in 0.5 ml of the same buffer. Cells were resuspended in 0.1 mlof Buffer A/10-cm plate and lysed by the addition of 0.5% (v/v) Nonidet

P-40, 0.5% (v/v) deoxycholate, and 0.1% (v/v) Triton X-100 and vortex-ing. Cell debris was removed by centrifugation in a microcentrifuge for

5 min at 4 °C, and the resultant supernatant was frozen in liquid N2.

 Measurement of Protein Synthesis—Cells were grown in six-wellplates and starved, as described above, prior to activation in the pres-

ence of 25 Ci/ml [35S]methionine, for the times described in the figure

legends. The medium was removed and cells washed in Buffer B (20 mM

Tris-HCl, pH 7.4, 0.134  M  NaCl, 1  M microcystin, 2 mM  benzamidine)

containing 5 mM unlabeled methionine, prior to lysis with 0.3  M  NaOH.Incorporation of radioactivity into protein was determined by precipi-tation with trichloroacetic acid.

 SDS-Polyacrylamide Gel Electrophoresis (SDS-PAGE), Vertical Slab Iso-electric Focusing (VSIEF), and Immunoblotting—One-dimensional

polyacrylamide gels and vertical slab iso-electric focusing gels were runas described (19), and proteins transferred to polyvinylidene difluoridemembrane. eIFE, eIF4G, and 4E-BP1 were detected with specific rabbitanti-peptide antisera as described previously (20) and in the individual

figure legends.m7 GTP-Sepharose Chromatography—For the isolation of eIF4E and

associated proteins, cell extracts of equal protein concentration were

subjected to m7GTP-Sepharose chromatography as described (19, 20).The beads were washed three times in Buffer A and bound proteineluted with either SDS-PAGE or VSIEF sample buffer, as indicated.

 Immunoprecipitation of eIF4G—Immunoprecipitation of eIF4G andassociated eIF4E from cell extracts of equal protein concentration was

as described previously (20). JNK/SAPK Kinase Assays—Solid-state protein kinase assays using 

the GST-c-Jun-(1–79) fusion protein were performed as described (23).

Briefly, samples of cell extracts (60   g) were mixed with 10   g of 

GST-c-Jun-(1–79) prebound to GSH-Sepharose. After 20 min at 30 °C,

the resin was recovered by centrifugation in a microcentrifuge, washedtwice in kinase assay buffer (50 mM Mops-KOH, pH 7.4, 20 mM MgCl

2,

40 mM -glycerophosphate, 1 M microcystin, 7 mM 2-mercaptoethanol,2 mM   benzamidine, 1 mM   phenylmethylsulfonyl fluoride, 1 mM

Na3 VO

4), and resuspended in 20  l of the same buffer containing 100

M [ -32P]ATP (1500 cpm/pmol). After 20 min at 30 °C, reactions wereterminated with SDS-PAGE sample buffer, followed by analysis by

SDS-PAGE and autoradiography.

RESULTS AND DISCUSSION

 Enhanced eIF4F Complex Formation in Vivo Can Occur In-

dependently of Enhanced 4E-BP1 Phosphorylation—eIF4E

plays a central role in the regulation of translation, with a

strong correlation between the phosphorylation of eIF4E,

eIF4F complex formation, and the rate of protein synthesis and

cell growth (reviewed in Ref. 1). However, the mechanism by

which phosphorylation of eIF4E at Ser-209 (25, 26) enhances

its activity is not understood. In addition to phosphorylation,

the activity of eIF4E can also be modulated by its availability to

participate in the initiation process, mediated by its interaction

with specific binding proteins, 4E-BP1 and 4E-BP2 (4, 15).

These studies have been facilitated by the use of rapamycin, an

inhibitor of the p70S6K  signaling pathway, which blocks cell

cycle progression (27), prevents the phosphorylation of 4E-BP1

in response to growth factors (1, 4, 16, 28), and inhibits cap

structure-dependent initiation of translation by the subsequent

inactivation of eIF4E (4, 15, 22). However, this may be a sim-

plistic view, as studies by Beretta et al. (22) have shown a lack

of temporal correlation between the inhibition of phosphoryla-

tion of 4E-BP1 and the inhibition of translation. In addition,

stabilization of the eIF4E/4E-BP1 complex did not prevent the

serum-stimulated phosphorylation of eIF4E  in vivo.

To examine this further, we have analyzed the correlation

between the phosphorylation of 4E-BP1, polysome formation,

eIF4F complex formation, and the activation of protein synthe-

sis in NIH 3T3 cells. At 4 h following serum stimulation (Fig.

1 A), rapamycin had only a small inhibitory effect (20–25%) on

the rate of total protein synthesis, under conditions where the

activation of p70S6K  was completely prevented (data not

shown). This inhibition was further increased to 30–35% by20 h. At early times after serum stimulation, rapamycin had

little effect on polysome formation (Fig. 1 B), even if the cells

had been preincubated with rapamycin prior to activation;

further incubation in the presence of rapamycin resulted in a

25–30% decrease in polysome formation (data not shown). We

have also examined the phosphorylation of 4E-BP1 and its

association with eIF4E. Multiple forms of 4E-BP1 can be dis-

tinguished on SDS-PAGE gels (Fig. 1C), with the    form iden-

tified as more highly phosphorylated than the     and    forms

(28). Serum stimulation resulted in the characteristic mobility

shift from the predominant    and    forms to the   form (lane 2

versus lane 1), an effect that was largely prevented by the

co-addition of rapamycin (lane 3). Isolation of eIF4E from the

unstimulated cells by m

7

GTP-Sepharose (lane 4) indicated, asexpected (1, 4) that the factor was mainly associated with the

less phosphorylated (   and  ) forms of 4E-BP1. As predicted

from the current models, enhanced phosphorylation of 4E-BP1

was concomitant with its release from eIF4E (lane 5 versus lane

 4), an effect largely prevented by rapamycin (lane 6). Preincu-

bation of cells with rapamycin for 30 min prior to activation

caused a greater accumulation of the    form of 4E-BP1, but did

not increase the level of 4E-BP1 associated with eIF4E above

that presented in lane 6.2 To determine the effect of rapamycin

on eIF4F complex formation, extracts were prepared following 

serum stimulation for 4 h in the absence or presence of rapa-

mycin prior to immunoprecipitation of eIF4G. The level of 

associated eIF4E was then assessed by SDS-PAGE and immu-

noblotting; the results of quantification of these data are pre-

sented in the figure legend. As shown in Fig. 1 D, serum treat-ment enhanced the recovery of eIF4E associated with eIF4G by

2.5-fold (lane 1 versus lane 3 and figure legend), indicative of a

stimulation of eIF4F complex assembly. Similar observations

have been reported for other cell types following activation of 

protein synthesis (reviewed in Ref. 1). Although rapamycin

prevented the phosphorylation of 4E-BP1 and stabilized its

association with eIF4E ( panel C,   lane 4 versus lane 6), the

recovery of eIF4E associated with eIF4G was enhanced by

2.3-fold. These data suggest that rapamycin did not prevent the

assembly of the eIF4F complex at 4 h following serum stimu-

lation, suggesting that enhanced eIF4F complex formation   in

2

S. J. Morley, unpublished data.

 eIF4E Phosphorylation during NIH 3T3 Cell Activation17888

Page 3: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 3/8

FIG. 1.   Rapamycin does not affect eIF4F complex formation in serum-stimulated NIH 3T3 cells.   Panel A, NIH 3T3 cells wereserum-starved for 48 h prior to the addition of either 50 n M rapamycin (rapa) or vehicle alone (rapa), 25  Ci/ml [35S] methionine, and serum(10%). Cells were harvested at the times indicated, and the incorporation of radioactive methionine into trichloroacetic acid-precipitable materialwas determined. The right-hand panel shows the effect of rapamycin expressed as the percentage of the control. These data are representative of those obtained in three separate experiments, and the error bars indicate the standard deviation from the mean.  Panel B, serum-starved cells wereincubated for 4 h in the absence or presence of FCS (10%), and in the absence or presence of 50 n M rapamycin, as indicated. Extracts were preparedin the presence of detergents, and equal amounts of protein were fractionated on sucrose density gradients, as described under “ExperimentalProcedures.” Sedimentation was from left to right, and an  arrow indicates the sedimentation of the 40 S ribosome.  Panel C, aliquots of extracts (40g) prepared as in  panel B  were either analyzed directly or subjected to m7GTP-Sepharose to isolate eIF4E and associated proteins, prior toSDS-PAGE and immunoblotting with antiserum specific to 4E-BP1 (a kind gift from J. Lawrence). The  -form of 4E-BP1 is the least phospho-rylated and the   -form is the most highly phosphorylated form of 4E-BP1. These data are representative of those obtained in three separateexperiments. Panel D, serum-starved cells were incubated in the absence or presence of 50 nM rapamycin or FCS (10%) for 4 h prior to preparationof extracts. Aliquots containing equal protein (50  g) were then subjected to immunoprecipitation with anti-eIF4G antiserum as described under“Experimental Procedures.” Isolated proteins were resolved by SDS-PAGE, and the recovery of eIF4G ( upper panel) and associated eIF4E (lower

 panel) was determined by immunoblotting and quantified by densitometric scanning. When expressed as the amount of eIF4G/eIF4E for each variable, they yielded the following: lane 1, 0.3 0.1 (S.D.,  n 3); lane 2, 0.4 0.1, (S.D., n 3); lane 3, 1.1 0.2 (S.D., n 3); lane 4, 0.9 0.1

(S.D., n

3).

 eIF4E Phosphorylation during NIH 3T3 Cell Activation   17889

Page 4: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 4/8

vivo   can occur independently of 4E-BP1 phosphorylation.

These findings are not consistent with the   in vitro   studies of 

Haghighat   et al.   (17), who reported that the interaction of 

4E-BP1 with eIF4E prevents the interaction of eIF4E with

eIF4G. Possible explanations for this are that either 4E-BP1 is

not present in excess of eIF4E in these cells or that there is a

population of eIF4E that is inaccessible to 4E-BP1 and is suf-

ficient to support enhanced protein synthesis at early times of 

activation. Indeed, even prolonged incubation with rapamycin

for 20 h resulted in only a 30–40% decrease in the amount of 

eIF4E associated with eIF4G in this system (data not shown).

 As suggested by Beretta  et al.   (22), these data may reflect a

slow rate of exchange of eIF4E through the eIF4F complex,

such that eIF4E is only able to interact with 4E-BP1 after it is

released from eIF4F.

The MAP Kinase Inhibitor, PD98059, but Not Rapamycin

 Prevents the Serum-induced Phosphorylation of eIF4E—We

have also examined the intracellular signaling pathways mod-

ulating the enhanced phosphorylation of 4E-BP1 and eIF4E,

the association of 4E-BP1 with eIF4E, and eIF4F complex

formation in response to serum stimulation. In addition to

rapamycin, we have used the following well characterized in-

hibitors: PD98059, to prevent activation of the classical MAP

kinase pathway (29); RO31-8220, which functions as a general

inhibitor of protein kinase C (30) but also stimulates the JNK/ 

SAPK signaling pathway (31); and SB203580, which is a spe-

cific inhibitor of the p38/RK MAP kinase (32). Fig. 2 A  shows

that there was a moderate increase in the rate of protein

synthesis over the initial 30 min following serum addition. As

shown in the Western blots in Fig. 2 B   (lane 2), enhanced

translation rates could be correlated with increased phospho-

rylation of 4E-BP1 and the dissociation of 4E-BP1 from eIF4E.

The concomitant shift of immunoreactive eIF4E to the upper

form on VSIEF indicated enhanced phosphorylation (19, 20).

Quantification of these data by densitometric scanning (see

figure legend) indicated that the percentage of total eIF4E in

the phosphorylated form was increased from 8% in the un-

stimulated cells to 40% following serum stimulation. In addi-tion, co-immunoprecipitation of eIF4E with an antiserum rec-

ognizing eIF4G, indicated a 2-fold increase in eIF4F complex

formation (see figure legend for quantifcation). In agreement

with published data, rapamycin did not suppress the effect of 

serum on the rate of translation (Fig. 2 A,  lane 4) or the phos-

phorylation of eIF4E (19–22), but largely prevented the phos-

phorylation of 4E-BP1 and stabilized its association with eIF4E

(Fig. 2 B,   lane 4). These data suggest that the association of 

eIF4E with 4E-BP1 does not necessarily prevent phosphoryla-

tion of eIF4E  in vivo. PD98059, which had little effect on the

rate of translation when added alone (Fig. 2 A,  lane 8) or fol-

lowing serum stimulation (Fig. 2 A,   lane 5), prevented activa-

tion of classical ERK2 MAP kinases (Ref. 29 and data not

shown) and attenuated the phosphorylation of eIF4E (Fig. 2 B,lane 5  and legend). However, it did not prevent the phospho-

rylation of 4E-BP1 or the dissociation of this protein from

eIF4E. Serum-stimulated association of eIF4E with eIF4G was

also unaffected by this inhibitor. These data are consistent

with published work, which shows that MAP kinase activation

is not required for increased phosphorylation of 4E-BP1 (16). In

addition, as shown for CHO.T cells (21), these data indicate

that signaling through the MAP kinase pathway is in part

responsible for mediating the phosphorylation of eIF4E. Con-

sistent with these findings is the observation that NIH 3T3

cells overexpressing MAP kinase kinase show an elevated ba-

sal level of eIF4E phosphorylation.2

 RO31-8220 Augments the Phosphorylation of eIF4E in Re-

sponse to Serum— A large number of studies have implicated

protein kinase C in regulation of eIF4E phosphorylation and

activity (reviewed in Ref. 1). Our data (Fig. 2 B, lane 6) confirms

other reports showing enhanced phosphorylation of eIF4E in

many cell types following treatment with the phorbol ester,

PMA. In NIH 3T3 cells, PMA caused a small but reproducible

increase in protein synthesis (Fig. 2 A,  lane 6). PMA treatment

also enhanced association of eIF4E with eIF4G by 3.3-fold (see

figure legend) but did not result in release of the binding 

protein from association with eIF4E (Fig. 2 B,   lane 6). The

widely used protein kinase C inhibitor, RO31-8220, was detri-

mental to basal rates of protein synthesis (data not shown) and

severely inhibited the response to serum and PMA (Fig. 2 A,

lanes 3   and   7 ). However, treatment of cells with RO31-8220

alone increased phosphorylation of eIF4E (data not shown),

and in combination with serum, augmented the enhanced phos-

phorylation of eIF4E (61% of total eIF4E in the phosphorylated

form; see legend for details) observed with serum alone (40% of 

eIF4E in the phosphorylated form; Fig. 2 B,  lane 3 versus lane

 2). This probably reflects the recent finding that RO31-8220

inhibits the expression of MAP kinase phosphatase, prolonging 

the activation of MAP kinase (31), a condition that could be

expected to enhance the phosphorylation of eIF4E. However,

since RO31-8220 is now known to activate the c-Jun N-termi-

nal kinase (JNK/SAPK) (31), our data also suggest the possi-

bility that this signaling pathway may have a role in the

enhanced phosphorylation of eIF4E.

 Activation of JNK/SAPK by Anisomycin Enhances eIF4E

 Phosphorylation—The three known groups of the MAP kinase

family include the classical MAP kinases (ERKs), stress-acti-

 vated kinases (JNK/SAPK) and p38 MAP kinase (p38/RK).

They are at the center of three distinct but closely related

phosphorylation cascades, which play a critical role in trans-

ducing extracellular signals into intracellular responses. The

archetypal MAP kinase pathway, activated by serum, growth

factors and mitogens, is stimulated in response to   ras-GTP

loading, activation of   raf   proto-oncogene, phosphorylation of 

mitogen-activated protein kinase kinase, which in turn phos-

phorylates and activates the MAP kinases, ERK1 and ERK2.In addition, this pathway is activated by G-protein signaling 

and phosphatidylinositol turnover (33–35). On the other hand,

cells respond to cellular stress agents by induction of two struc-

turally related but distinct pathways, JNK/SAPK and p38/RK 

(34). JNK/SAPK is activated by a mitogen-activated protein

kinase kinase-like kinase, SEK1 (MKK4, JNKK), while p38/RK 

is phosphorylated by related kinases MKK3 and MKK6, which

are themselves part of an ill-defined, overlapping signaling 

cascade (33–35). Study of these signaling cascades is compli-

cated by cross-talk between them, but it is clear that JNK/ 

SAPK and p38/RK activation culminates in the activation of 

transcription factors, enhanced expression of the immediate-

early genes c- fos  and c- jun  and hence in the regulation of cell

growth and differentiation (33–36).To investigate the potential role of the JNK/SAPK and

p38/RK MAP kinase signaling pathways in the phosphoryla-

tion of eIF4E and its association with 4E-BP1, we have used

anisomycin, which strongly activates JNK/SAPK in numerous

cell types (33–37) in conjunction with serum and the inhibitors

listed above. As shown in Fig. 3 A, serum enhanced the amount

of total eIF4E in the phosphorylated form from 10% in control

cells to 47% (see legend for details), promoted the dissociation

of 4E-BP1 from eIF4E, but did not greatly increase JNK/SAPK 

activity in NIH 3T3 cells (lane 2 versus lane 1). The apparent

less complete serum-stimulated dissociation of 4E-BP1 from

eIF4E than presented in Fig. 2 reflects the use of a more

sensitive antiserum in this experiment (see figure legend).

Serum-stimulated phosphorylation of eIF4E was insensitive to

 eIF4E Phosphorylation during NIH 3T3 Cell Activation17890

Page 5: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 5/8

the presence of SB203580 (lane 4); however, treatment with

SB203580 alone resulted in the complete dephosphoryation of 

eIF4E (lane 3; quantified in figure legend). These data suggest

that either p38/RK MAP kinase is involved in the negative

regulation of an eIF4E phosphatase or else it is partially acti-

 vated in serum-starved cells. Anisomycin, which activated the

JNK/SAPK signaling pathway (Fig. 3 A,  lane 5), enhanced the

level of total eIF4E in the phosphorylated form to 50% and

promoted the dissociation of the 4E-BP1/eIF4E complex. Sim-

ilar data were obtained when levels of anisomycin insufficient

to inhibit the elongation phase of translation were employed

(data not shown). Interestingly, in contrast to serum, anisomy-

cin-induced phosphorylation of eIF4E was sensitive to rapamy-

FIG. 2.  Inhibition of the MAP kinase but not the p70S6K  signal-ing pathway prevents the phosphorylation of eIF4E but notformation of the eIF4F complex.  Panel A, serum-starved cells wereincubated in six-well plates in the absence or presence of PD98059 (50M), rapamycin (50 nM), or RO31-8220 (50  M) for 15 min, as indicated.

Cells were then incubated with 25  Ci/ml [35S]methionine, in the ab-sence or presence of FCS (10%) or PMA (50 nM) for 15 min, beforeharvesting to determine the incorporation of label into protein. Theexperiment was carried out three times, each in triplicate, and the errorbars indicate the standard deviation from the mean.   Panel B, cellextracts of equal protein concentration prepared as in   panel A   wereanalyzed by SDS-PAGE and the phosphorylation status on 4E-BP1determined by immunoblotting (antiserum kindly provided by J. Law-rence); isolation of eIF4E and associated proteins was determined bym7GTP-Sepharose chromatography and the amount of 4E-BP1 associ-ated with eIF4E determined by immunoblotting (m7GTP-Sepharoseeluate). Isolation of eIF4E was by m7GTP-Sepharose chromatography,and analysis of its phosphorylation status was by VSIEF. Densitometricscanning of the level of total eIF4E in the phosphorylated form yieldedthe following:  lane 1, 8%;  lane 2, 40%;  lane 3, 61%;  lane 4, 42%;  lane 5,19%;  lane 6, 41%; lane 7 , 39%; these data are from a single experimentbut are representative of those obtained in five separate experiments.eIF4G was immunoprecipitated from extracts as described and theamount of associated eIF4E by visualized by immunoblotting ( IP).Quantification of these data, as described in Fig. 1 D, yielded the fol-lowing: lane 1, 0.15; lane 2, 0.30;  lane 3, 0.35; lane 4, 0.40;  lane 5, 0.35;lane 6, 0.50. These data are from a single experiment but are repre-sentative of those obtained in three separate experiments.

FIG. 3. Phosphorylation of eIF4E is stimulated by activation of the JNK/SAPK signaling pathway. Panel A, serum-starved NIH 3T3cells were incubated with SB203580 (10   M), rapamycin (50 nM), orPD98059 (50   M) for 30 min prior to the addition of FCS (10%) oranisomycin (10   g/ml) for 15 min, as indicated. Cell extracts wereprepared and equal amounts of protein subjected to m7GTP-Sepharoseto isolate eIF4E and associated proteins. The   upper panel  shows theresult of VSIEF analysis of recovered eIF4E; quantification of theamount of total eIF4E in the phosphorylated form by densitometricscanning yielded the following: lane 1, 10%; lane 2, 47%; lane 3, 0%; lane

 4, 45%;  lane 5, 50%;  lane 6, 25%;  lane 7 , 2%;  lane 8, 20%;  lane 9, 45%.The middle panel  shows Western blotting of the level of 4E-BP1 asso-ciated with eIF4E following isolation by m7GTP-Sepharose chromatog-raphy (antiserum kindly provided by R. Denton). The lower panel showsthe activity of JNK/SAPK present in cell extracts assayed using GST-c-Jun-(1–79) as ligand and substrate, as described under “ExperimentalProcedures.” These data are from a single experiment but are repre-sentative of those obtained in three separate experiments.   Panel B,serum-starved NIH 3T3 cells were incubated in the absence or presenceof rapamycin (50 nM) or PD98059 (50 nM) for 30 min, prior to theaddition of FCS (10%) or anisomycin (10 g/ml) for 15 min. PMA (50 nM)was added for 15 min, as indicated, and cell extracts were prepared. Thesteady state phosphorylation status of eIF4E was determined by VSIEF

(upper panel) and quantification of the amount of total eIF4E in thephosphorylated yielded the following:  lane 1, 7.7%;  lane 2, 47.0%;  lane 3, 45.0%;  lane 4, 13.0%;  lane 5, 41.0%;  lane 6, 7.0%;  lane 7 , 45.0%. Todetermine the level of eIF4E associated with ribosomes, extracts werecentrifuged at 100,000 rpm in a Beckman TL100 centrifuge for 30 minat 4 °C and the isolated ribosomes resuspended in Buffer A, with theaddition of 0.5  M  KCl and 5 mM MgCl2. The salt-washed ribosomes werere-isolated by centrifugation and the supernatant diluted to 100 mM

KCl prior to isolation of eIF4E by m7GTP-Sepharose and analysis bySDS-PAGE and immunoblotting (lower panel). These data are from asingle experiment but are representative of those obtained in threeseparate experiments.

 eIF4E Phosphorylation during NIH 3T3 Cell Activation   17891

Page 6: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 6/8

cin (lane 6) and SB203580 (lane 7 ), but not PD98059 (lane 9;

see legend for quantification). These data indicate that more

than one family of MAP kinase is involved in regulating the

phosphorylation status of eIF4E in NIH 3T3 cells.

Previously, we have shown that in primary T cells and fol-

lowing meiotic maturation of  Xenopus oocytes, the activation of 

protein synthesis can be correlated with enhanced recruitment

of eIF4E to the ribosome (19, 20). To analyze the potential role

for increased phosphorylation of eIF4E in promoting interac-

tion of the factor with ribosomes, serum-starved cells were

stimulated with serum in the absence or presence of the inhib-

itors described above, and the level of ribosome-associated

eIF4E visualized by immunoblotting. Fig. 3 B   shows that, rel-

ative to the unstimulated cells (lane 1, 7.7% of eIF4E in the

phosphorylated form), both serum (lane 2, 47% of eIF4E in the

phosphorylated form) and PMA (lane 7 , 45% of eIF4E in the

phosphorylated form) enhanced the phosphorylation of total

eIF4E (upper panel) and increased the recovery of eIF4E on

ribosomes (lower panel). Rapamycin (lane 5) and PD98059

(lane 6) had little effect on the recruitment of eIF4E to the

ribosome in response to serum stimulation, although PD98059

did prevent the enhancement of phosphorylation of eIF4E.

Further fractionation of ribosomes by sucrose density gradient

analysis showed that serum treatment enhanced binding of eIF4E to the 40 S ribosomal subunit; VSIEF and immunoblot

analysis of this population of eIF4E indicated that, as seen with

the reticulocyte lysate (38), it consisted of a population includ-

ing both phosphorylated and non-phosphorylated forms (data

not shown). In contrast, anisomycin treatment enhanced the

level of eIF4E in the phosphorylated form to 45% without

promoting its association with the ribosome (lane 3). Interest-

ingly, while rapamycin prevented the anisomycin-induced

phosphorylation of eIF4E (lane 4; see legend for details), it

resulted in recruitment of eIF4E to the ribosome, albeit to a

lower level than observed with serum (lane 2). The reasons for

this are unclear at this time.

The simplest explanation for these observations would be

that serum and/or anisomycin activates an eIF4E kinase via aMAP kinase signaling pathway. Anisomycin, but not serum,

induced the activation of p38 MAP kinase, the activation of 

MAPKAPK-2, and the phosphorylation of a peptide substrate

designed after a phosphorylation site in HSP 27 (39) (data not

shown). A role for MAP kinases in mediating the enhanced

phosphorylation of eIF4E have been proposed during insulin

stimulation of CHO.T cells (21), in mediating the activation of 

a protamine kinase which can phosphorylate eIF4E   in vitro

(40–42), and can be inferred by the finding that lipopolysac-

charide (which stimulates p38 MAP kinase activity; Refs. 33–

35), enhanced the phosphorylation of eIF4E in macrophages

(43). However, neither MAP kinase itself (1), p38 MAP kinase,2

nor downstream targets of MAP kinase, such as MAPKAPK-1

or MAPKAPK-2 (21) can directly phosphorylate eIF4E  in vitro,

although each was active when assessed using characterizedsubstrates. Recent studies with 3pK (MAPKAPK-3), a kinase

targeted by MAP kinase pathways (39, 44), have shown that

this kinase will directly phosphorylate eIF4E   in vitro   when

immunoprecipitated from arsenite-stimulated transfected hu-

man 293 cells (Institut fur Medizinische Strahlenkunde und

Zellforschung, Wurzburg, Germany).3 Further work is required

to determine whether 3pK is a physiological eIF4E kinase and

if it plays any role in the enhanced phosphorylation of eIF4E in

NIH 3T3 cells under any conditions described above.

Studies with numerous cell types (1, 45, 46) have suggested

a role for regulated phosphatase activity in the enhanced phos-

phorylation of eIF4E. In NIH3T3 cells this is also likely as the

phosphatase inhibitor, okadaic acid, enhanced the phosphoryl-

ation of eIF4E to levels seen with serum (data not shown).

 Arsenite, which activates p38/RK and JNK/SAPK, also en-

hanced the phosphorylation of eIF4E in these cells.2 However,

as arsenite inhibits the activity of a constitutive dual-specific-

ity phosphatase in human cells (47), further work is required to

determine whether this phosphatase targets eIF4E and is re-

sponsible for the dephosphorylation of eIF4E observed with

SB203580 (Fig. 3 A, lane 3). Therefore, as described for human

cells (48), our studies indicate that the phosphorylation of 

eIF4E and 4E-BP1 can be regulated independently and that

each may play a direct role in translational initiation in vivo.

 Acknowledgments— Antiserum to 4E-BP1 (PHAS-I) was kindly pro- vided by Dr. J. Lawrence, Jr. and Prof. R. Denton, and the plasmidencoding the glutathione   S-transferase fusion protein GST-c-Jun-(1–79) was provided by Dr. M. Karin. We thank Dr. J. Kay for rapamycin,Parke-Davis for PD098059, The Roche Research Center for RO31-8220,and SmithKline Beecham for SB203580.

REFERENCES

1. Morley, S. J. (1996) in   Protein Phosphorylation in Cell Growth Regulation(Clemens, M. J., ed) pp. 197–224, Harwood Academic Publishers, Amsterdam

2. Pain, V. M. (1996) Eur. J. Biochem.  236,  747–771

3. Sonenberg, N. (1994) Biochimie  76,  839–8464. Sonenberg, N.(1996)in Translational Control (Hershey,J. W.B., Mathews,M.

B., and Sonenberg, N., eds) pp. 245–269, Cold Spring Harbor Laboratory,Cold Spring Harbor, NY 

5. Hershey, J. W. B. (1989) J. Biol. Chem.  264,  20823–208266. Proud, C. G. (1992) Curr. Top. Cell. Regul.  32,  243–2697. Rhoads, R. E. (1993) J. Biol. Chem.  268,  3017–30208. Pause, A., Methot, N., Svitkin, Y., Merrick, W. C., and Sonenberg, N. (1994)

 EMBO J.  13,  1205–12159. Lamphear, B. J., Kirchweger, R., Skern, T., and Rhoads, R. E. (1995) J. Biol.

Chem. 270,  21975–2198310. Ohlmann, T., Rau, M., Pain, V. M., and Morley, S. J. (1996)   EMBO J.   15,

1371–138211. Mader, S., Lee, H., Pause, A., and Sonenberg, N. (1995)   Mol. Cell. Biol.  15,

4990–499712. Minich, B., Balasta, M. L., Goss, D. J., and Rhoads, R. E. (1994)  Proc. Natl.

 Acad. Sci. U. S. A. 91,  7668–767213. Bu, X., Haas, D. W., and Hagedorn, C. H. (1993) J. Biol. Chem.  268,  4975–7814. Morley, S. J., Rau, M., Kay, J. E., and Pain, V. M. (1993) Eur. J. Biochem.  218,

39–48

15. Pause, A, Belsham, G. J., Gingras, A.-C., Donze, O., Lin, T.-A., Lawrence, J. C.,and Sonenberg, N. (1994)  Nature  371,  762–767

16. Von Manteuffel, S. R., Gingras, A.-C., Ming, X.-F., Sonenberg, N., and Thomas,G. (1996) Proc. Natl. Acad. Sci. U. S. A.  93,   4076–4048

17. Haghighat, A., Mader, S., Pause, A., and Sonenberg, N. (1995) EMBO J.  14,5701–5709

18. Whalen, S. G., Gingras, A.-C., Amankwa, L., Mader, S., Branton, P. E., Aebersold, R., and Sonenberg, N. (1996) J. Biol. Chem.  271,  11831–11837

19. Morley, S. J., and Pain, V. M. (1995) Biochem. J.  312,  627–63720. Morley, S. J., and Pain, V. M. (1995) J. Cell Sci.  108,  1751–176021. Flynn, A., and Proud, C. G. (1996) FEBS Lett.  389,  162–16622. Beretta, L., Gingras, A.-C., Svitkin, Y. V., Hall, M. N., and Sonenberg, N.

(1996) EMBO J.  15,  658–66423. Moxham, C. M., Tabrizchi, A., Davis, R. J., and Malbon, C. C. (1996)  J. Biol.

Chem.  271,  30765–3077324. Deleted in proof 25. Joshi, B., Cai, A.-L., Keiper, B. D., Minich, W. B., Mendez, R., Beach, C. M.,

Stepinski, J., Stolarski, R., Darzynkiewicz, E., and Rhoads, R. E. (1995) J. Biol. Chem.  270,  14597–14603

26. Flynn, A., and Proud, C. G. (1995) J. Biol. Chem.  27 0,  21684–21688

27. Brown, E. J., and Schneider, S. L. (1996) Cell  86,  517–52028. Graves, L. M., Bornfeldt, K. E., Argast, G. M., Krebs, E. G., Kong, X., Lin,

T.-A., and Lawrence, J. C. (1995)   Proc. Natl. Acad. Sci. U. S. A.   92,7222–7226

29. Pang, L., Sawada, T., Decker, S. J., and Saltiel, A. R. (1995) J. Biol. Chem. 270,13585–13588

30. Nixon, J. S., Bishop, J., Bradshaw, D., Davis, P. D., Hill, C. H., Elliot, L. H.,Kumar, H., Lawton, G., Lewis, E. J., Mulqueen, M., Westmacott, D., Wad-sworth, J., and Wilkinson, S. E. (1992)  Biochem. Soc. Trans.  20,  419–425

31. Beltman, J., McCormick, F., and Cook, S. J. (1996)   J. Biol. Chem.   271,27018–27024

32. Cuenda, A., Rouse, J., Doza, Y. N., Meier, R., Cohen, P., Gallagher, T. F., Young, P. R., and Lee, J. C. (1995) FEBS Lett.  364,  229–233

33. Pelech, S. (1996) Curr. Biol.  6,  551–55434. Kyriakis, J. M., and Avruch, J. (1996)   BioEssays  18,  567–57735. Davis, R. J. (1994) Trends Biochem. Sci.  19,  470–47336. Shu, J., Hitomi, M., and Stacey, D. (1996)   Oncogene  13,  2421–243037. Kardanilou, E., Zhelev, N., Hazzalin, C. A., and Mahadevan, L. C. (1994) Mol.

Cell. Biol.  14,  1066–107438. Rau, M., Ohlmann, T., Morley, S. J., and Pain, V. M. (1996) J. Biol. Chem. 271,

8983–89903

S. Ludwig, S. J. Morley, and L. McKendrick, unpublished data.

 eIF4E Phosphorylation during NIH 3T3 Cell Activation17892

Page 7: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 7/8

39. Clifton, A. D., Young, P. R., and Cohen, P. (1996)  FEBS Lett.  392,  209–21440. Amick, G. D., and Damuni, Z. (1992) Biochem. Biophys. Res. Commun.  183,

431–43741. Reddy, S. A. G., Guo, H., Tarun, S. Z., Jr., and Damuni, Z. (1993) J. Biol. Chem.

268, 15298 –1530442. Makkinje, A., Xiong, H., Li, M., and Damuni, Z. (1995) J. Biol. Chem.  270,

14824–1482843. Haas, D. W., Shepherd, V. L., and Hagedorn, C. H. (1992) Second Messengers

 Phosphoproteins  14,  163–171

44. Ludwig, S., Engel, K., Hoffmeyer, A., Sithanandam, G., Neufeld, B., Palm, D.,Gaestel, M., and Rapp, U. (1996)  Mol. Cell. Biol.  16,  6687–6697

45. Donaldson, R. W., Hagedorn, C. H., and Cohen, S. (1991) J. Biol. Chem.  266,3162–3166

46. Kleijn, M., Voorma, H. O., and Thomas, A. A. M. (1996)  J. Cell. Biochem.  59,443–452

47. Cavigelli, M., Li, W. W., Lin, A., Su, B., Yoshioka, K., and Karin, M. (1996) EMBO J.  15,   6269–6279

48. Feigenblum, D., and Schneider, R. J. (1996) Mol. Cell. Biol.  16,  5450–5457

 eIF4E Phosphorylation during NIH 3T3 Cell Activation   17893

Page 8: J. Biol. Chem.-1997-Morley-17887-93

7/21/2019 J. Biol. Chem.-1997-Morley-17887-93

http://slidepdf.com/reader/full/j-biol-chem-1997-morley-17887-93 8/8

Simon J. Morley and Linda McKendrick  Factor 4E in NIH 3T3 CellsEnhanced Phosphorylation of InitiationProtein Kinase Signaling Pathways in theKinase and p38/RK Mitogen-activatedInvolvement of Stress-activated ProteinGENETICS:SYNTHESIS, AND MOLECULARNUCLEIC ACIDS, PROTEIN

doi: 10.1074/jbc.272.28.17887 1997, 272:17887-17893.J. Biol. Chem.

http://www.jbc.org/content/272/28/17887Access the most updated version of this article at

.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the 

Alerts: 

When a correction for this article is posted•When this article is cited•

to choose from all of JBC's e-mail alertsClick here

 http://www.jbc.org/content/272/28/17887.full.html#ref-list-1This article cites 45 references, 22 of which can be accessed free at