You are on page 1of 9

CELL BIOLOGY AND METABOLISM:

p38 Mitogen-activated Protein Kinase


Pathway Promotes Skeletal Muscle
Differentiation: PARTICIPATION OF
THE MEF2C TRANSCRIPTION
FACTOR

Anna Zetser, Eran Gredinger and Eyal Bengal


J. Biol. Chem. 1999, 274:5193-5200.
doi: 10.1074/jbc.274.8.5193

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


Access the most updated version of this article at http://www.jbc.org/content/274/8/5193

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

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

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

This article cites 44 references, 32 of which can be accessed free at


http://www.jbc.org/content/274/8/5193.full.html#ref-list-1
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 274, No. 8, Issue of February 19, pp. 5193–5200, 1999
© 1999 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

p38 Mitogen-activated Protein Kinase Pathway Promotes Skeletal


Muscle Differentiation
PARTICIPATION OF THE MEF2C TRANSCRIPTION FACTOR*

(Received for publication, June 10, 1998, and in revised form, November 6, 1998)

Anna Zetser‡, Eran Gredinger‡, and Eyal Bengal§


From the Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine,
Technion-Israel Institute of Technology, Haifa 31096, Israel

Differentiation of muscle cells is regulated by extra- activation of phosphatidylinositol 3-kinases and mitogen-acti-
cellular growth factors that transmit largely unknown vated protein kinases (MAPK)1 via tyrosine kinase receptors
signals into the cells. Some of these growth factors in- within many cell types including muscle (5, 6). One MAPK
duce mitogen-activated protein kinase (MAPK) cascades induced by insulin is p38 MAPK (6). p38 is also activated by
within muscle cells. In this work we show that the ki- exposure of cells to environmental stress or by treatment of
nase activity of p38 MAPK is induced early during ter- cells with pro-inflammatory cytokines (7, 8). The affectors reg-
minal differentiation of L8 cells. Addition of a specific ulating p38 are only partly explored. However, the direct in-
p38 inhibitor SB 203580 to myoblasts blocked their fu- tracellular activators of p38 are MKK3 and MKK6 (MAPK

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


sion to multinucleated myotubes and prevented the ex- kinase) (9, 10). The role of p38 in skeletal muscle differentia-
pression of MyoD and MEF2 family members and myo- tion is not known. Several recent findings suggest that p38
sin light chain 2. The expression of MKK6, a direct
MAPK may be involved in skeletal muscle differentiation: 1)
activator of p38, or of p38 itself enhanced the activity of
Transcripts of MKK6 gene are most abundant in skeletal mus-
MyoD in converting 10T1/2 fibroblasts to muscle,
cle (10). 2) Insulin that promotes skeletal muscle differentia-
whereas treatment with SB 203580 inhibited MyoD. Sev-
eral lines of evidence suggesting that the involvement of tion also induces p38 activity (6). 3) p38 pathway regulates the
p38 in MyoD activity is mediated via its co-activator expression of glucose transporters in skeletal muscle cells (11).
MEF2C, a known substrate of p38, are presented. In 4) p38 activates directly the transcription factor MEF2C in
these experiments we show that MEF2C protein and inflammation (12). Together with the MyoD family, members of
MEF2-binding sites are necessary for the p38 MAPK the MEF2 family are necessary for the differentiation of myo-
pathway to regulate the transcription of muscle crea- blasts (13). 5) p38 pathway plays a role in specific gene expres-
tine kinase reporter gene. Our results indicate that the sion and cell hypertrophy of the related cardiac muscle lineage
p38 MAPK pathway promotes skeletal muscle differen- (14). In this report we show that 1) the p38 pathway plays an
tiation at least in part via activation of MEF2C. essential role in the in vitro differentiation of myoblasts and 2)
that this role may be mediated via the MEF2C transcription
factor.
The development of skeletal muscle is a multistep process in
EXPERIMENTAL PROCEDURES
which pluripotent mesodermal cells give rise to myoblasts that
Materials
subsequently withdraw from the cell cycle and differentiate
into myotubes. These stages are driven by the expression and SB 203580 was a product of Calbiochem. Goat polyclonal antibody
raised against amino acids 341–360 of mouse p38 was obtained from
activity of myogenic transcription factors from the MyoD fam-
Santa Cruz Biotechnology. Rabbit polyclonal antibody that detects p38
ily (1). Two members of the MyoD family, Myf5 and MyoD, are MAPK only when activated by dual phosphorylation at Thr180 and
expressed in dividing myoblasts that are committed to the Tyr182 was purchased from New England Biolabs. Antibodies to MEF2A
myogenic lineage. However, these proteins are not functional in and C proteins and to GAL4 (amino acids 94 –147) were purchased from
myoblasts, and their activities are induced only at a subse- Santa Cruz Biotechnology. Protein A-Sepharose was supplied by Sigma.
quent stage to allow withdrawal from the cell cycle and termi- Plasmids
nal differentiation (2). Induction of the activity of MyoD and
pEMSV-MyoD was described by Tapscott et al. (15). The 4R-tk-CAT
Myf5 proteins may be a result of the activity of extracellular
reporter gene was described by Weintraub et al. (16). pe1AT-CAT and
growth factors like insulin and insulin-like growth factors p(1enh110)-80MCK-CAT were described by Buskin and Hauschka (17).
known to promote terminal differentiation of myoblasts (3, 4). For MEF2– 80MCK-CAT, three copies of a double-stranded oligonucleo-
Insulin and insulin-like growth factors are involved in the tide of the MEF2 site (30 base pairs) from the MCK enhancer were
inserted upstream to a minimal MCK promoter (280 to 17 relative to
the transcription start site). pGEX-ATF2 plasmid was a gift from Dr.
* This work was supported by a United States-Israel Binational Ami Aronheim and Dr. Michael Karin. The wild type (MKK6) and
Foundation grant, by an Israel Cancer Association grant, and by funds activated (MKK6b(E)) alleles of MKK6 were described by Han et al.
from the Rappaport Foundation for Medical Research and the Founda- (10). The MEF2C expression vector (pCDNAI-MEF2C) was a generous
tion for Promotion of Research in the Technion, Israel Institute of gift of Dr. E. Olson (18). The inactive form of MEF2C was generated by
Technology. The costs of publication of this article were defrayed in part deleting a BglII fragment that removed amino acids 202–321 of
by the payment of page charges. This article must therefore be hereby
marked “advertisement” in accordance with 18 U.S.C. Section 1734
1
solely to indicate this fact. The abbreviations used are: MAPK, mitogen-activated protein ki-
‡ These authors contributed equally to the work. nase; MCK, muscle creatine kinase; ER, estrogen receptor; ERK, extra-
§ To whom correspondence should be addressed: Dept. of Biochemis- cellular signal-regulated protein kinase; DM, differentiation medium;
try, Faculty of Medicine, Technion-Israel Inst. of Technology, P.O. Box CAT, chloramphenicol acetyltransferase; GST, glutathione S-transfer-
9649, Haifa 31096, Israel. Tel.: 972-4-8295-287; Fax: 972-4-8535-773; ase; PAGE, polyacrylamide gel electrophoresis; MHC, myosin heavy
E-mail: Bengal@tx.technion.ac.il. chain.

This paper is available on line at http://www.jbc.org 5193


5194 Role of p38 Pathway in Myogenesis
MEF2C. GAL4-MEF2C constructs were described by Han et al. (12), 1:100 and anti-phospho-p38 antibody (New England Biolabs) 1:100.
and GAL4-MyoD constructs were described by Weintraub et al. (19). Proteins were visualized using the enhanced chemiluminescence kit of
pGEX-MEF2C was constructed by inserting a polymerase chain reac- Amersham Pharmacia Biotech.
tion fragment encoding amino acids 128 – 467 of MEF2C into the
pGEX-2T vector.
Metabolic Labeling of Cells and Immunoprecipitation of MEF2C
Cell Culture Labeling with [35S]Methionine—Cells were incubated in methionine-
L8 cells were a gift of Dr. David Yaffe (20). 10T1/2 cells were obtained free Dulbecco’s modified Eagle’s medium and dialyzed calf serum for 40
from ATCC. 10T1/2 cells that expressed the MyoD-estrogen receptor min and then incubated with 100 mCi/ml [35S]methionine for 3 h before
(ER) chimera protein were described by Hollenberg et al. (21). Cell lines proteins were extracted in RIPA buffer (50 mM Tris, pH 7.9, 150 mM
were maintained in Dulbecco’s modified Eagle’s medium supplemented NaCl, 1% Nonidet P-40, 0.1% SDS, 0.5 mM dithiothreitol, 0.1 mM
with 15% calf serum (Hyclone), penicillin, and streptomycin (growth Na3VO4, 2 mg/ml leupeptin, 20 mM p-nitrophenyl phosphate, and 100
medium). To induce differentiation, we used Dulbecco’s modified Ea- mg/ml phenylmethylsulfonyl fluoride).
gle’s medium supplemented with 10 mg of insulin/ml and 10 mg of Labeling with [32P]Orthophosphate—L8 cells were incubated in Dul-
transferrin/ml (differentiation medium, DM). Differentiation of 10T1/2 becco’s modified Eagle’s medium without sodium phosphate to which
cells that expressed MyoD-ER protein was induced by the addition of [32P]orthophosphate was added at 0.75 mCi/ml for 3 h before proteins
DM that contained 1027 M estradiol. were extracted in RIPA buffer.
Immunoprecipitation—Equal amounts of labeled proteins were incu-
Growth of Cells in the Presence of SB 203580 bated with antibody for 2 h and then for an additional hour with protein
SB 203580 was dissolved in Me2SO to a concentration of 10 mM and A-Sepharose. The protein A-Sepharose beads were washed four times
was added directly to the differentiation medium to a final concentra- with RIPA containing 0.5 M NaCl and once in RIPA.
tion of 10 –20 mM as indicated. Control cells were incubated with the
RESULTS
same volumes of Me2SO without SB 203580. The medium was replaced
every 12 h with medium containing fresh SB 203580. p38 Activity Is Induced during Differentiation of L8 Cells—
The in vitro kinase activity of p38 was studied using extracts of

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


Transfections
L8 muscle cells from different stages of differentiation. Kinase
Transfections were performed by calcium phosphate precipitation as
activity of p38 was analyzed by immunoprecipitation of p38,
described (22). Cells in 6-cm TC dishes (Corning) were transfected for
12 h with a total amount of 10 mg of the following plasmid DNA: 1 mg of which was used to phosphorylate its substrate, ATF2 (Fig. 1A).
pCMV-LacZ, 3 mg of CAT reporter gene, 3 mg of MyoD expression The kinase activity of p38 was present in dividing myoblasts
plasmid, 3 mg of MEF2C expression vector, and 3 mg of expression and was gradually induced during the differentiation to
vector of wt MKK6 or activated form of MKK6b (MKK6b(E)). Following multinucleated myotubes. Kinase activity was specific; it was
transfection, the medium was replaced with either growth medium or induced by UV irradiation and inhibited by the p38-specific
DM for another 24 – 48 h. The efficiency of transfections were tested in
inhibitor SB 203580 added to L8 cells (Fig. 1A). The amount of
soluble 5-bromo-4-chloro-3-indolyl b-D-galactopyranoside assays as de-
scribed (23), and the amount of extracts used for the CAT assays were the phosphorylated form of p38 that reflects the active kinase
adjusted accordingly (24). was studied in L8 cells. An antibody that recognizes the active
dual phosphorylated form of p38 was used with the same ex-
Immunohistochemical Staining tracts in a Western analysis. Phosphorylated forms of p38
Cells were fixed and immunostained as described (25). The primary accumulated during the differentiation of cells in DM (Fig. 1B).
antibodies used were monoclonal anti-MyoD (5.8A) and polyclonal anti- The level of total p38 protein in differentiating cells was not
MHC (Sigma). The immunochemically stained cells were viewed at
constant; however, the calculated percentage of phosphorylated
3200 magnification in a fluorescence microscope (Olympus, model
BX50). p38 in cells growing in DM for 48 h was 2–3-fold higher than
that of dividing myoblasts (Fig. 1B). Next, we assayed p38
In Vitro Kinase Assays kinase activity with a second substrate, GST-MEF2C. p38 was
Expression of GST Fusion Proteins—The GST-ATF2 and GST- recently demonstrated to associate and directly phosphorylate
MEF2C (128 – 467) proteins were expressed from the bacterial expres- MEF2C (12). In addition, MEF2C is an essential transcription
sion vectors in BL21 strain of Escherichia coli and purified from ex- activator of muscle-specific genes (13). Phosphorylation of GST-
tracts on glutathione beads as described (26).
MEF2C substrate was increased during the differentiation of
Preparation of Cell Extracts—Cell extracts were prepared as de-
scribed (25). L8 myoblasts to multinucleated myotubes (Fig. 1C). p38 was
In Vitro Kinase Assay for p38 —Cells were extracted in lysis buffer probably the kinase that phosphorylated GST-MEF2C because
(described above). Equal amounts of protein from each time point of kinase activity was significantly inhibited in cells treated with
differentiation were rotated with 6 ml of anti-p38 antibody (Santa Cruz) SB 203580 (Fig. 1C, lanes 3 and 4). The increased activity of
for 2.5 h at 4 °C. The immunoprecipitation procedure and the kinase p38 during muscle differentiation suggests that it might have a
assay were performed as described for ERK (25). However, the sub-
role in this process. Four isoforms of p38 (a, b, g, and d) are
strate in the present assay was GST-ATF2 (20 mg).
In Vitro Kinase Activity of p38 Using GST-MEF2C as a Substrate— presently known. To study the relative contributions of p38
Equal amounts of cell extracts were collected at different times after the isoforms in the phosphorylation of MEF2C, each isoform was
initiation of cell differentiation and were mixed with GSH-agarose immunodepleted from muscle extracts. Differential depletion of
beads to which GST or GST-MEF2C proteins were bound. The mixtures p38 isoforms affected the phosphorylation of MEF2C (Fig. 1C,
were processed, and the kinase assay was performed as described by lanes 6 –11). p38a contributed most of p38 kinase activity (40%
Hibi and colleagues for the JNK kinase assay (26). Results were quan-
inhibition), whereas p38b and d were less active (25 and 10%
tified by PhosphorImager.
inhibition, respectively). Depletion of p38g did not inhibit the
RNA Analysis phosphorylation of MEF2C (Fig. 1C, lane 10).
RNA was extracted and analyzed by Western blotting as described Treatment of L8 Cells with SB 203580 Inhibits Their Differ-
(25). Blots were hybridized with probes for MEF2C (pEMSV-MEF2C), entiation—To evaluate the role of p38 in myogenesis, we used
MyoD (pEMSV-MyoD), myogenin (pEMSV-myogenin), MLC2 (PV- the p38 inhibitor SB 203580 (27) that was added to L8 cells at
ZLC2), p21 (pCDNA-Waf1), and GAPDH (pMGAP). concentrations that blocked most of the p38 activity (Fig. 1A).
Western Analysis The inhibitor was added concomitantly with the induction of
differentiation (differentiation medium, see “Experimental
Cells were lyzed as described for the kinase assays, and equal
amounts of extracted proteins were loaded and separated by SDS- Procedures”). After 36 h in that medium, cells not treated with
PAGE and transferred to nitrocellulose filters. Immunoblotting was the drug fused to form developed myotubes, whereas cells
conducted with the following two antibodies: anti-p38 (Santa Cruz) treated with the drug did not form myotubes (Fig. 2). Moreover,
Role of p38 Pathway in Myogenesis 5195

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


FIG. 2. The p38 inhibitor, SB 203580, prevents the fusion of L8
myoblasts to multinucleated myotubes. The p38 inhibitor, SB
203580 (20 mM), was added to L8 cells immediately after the addition of
FIG. 1. p38 kinase activity is induced during differentiation of DM when cells were about 80% confluent. SB 203580 was replaced with
L8 cells. A, the in vitro kinase activity of p38 was studied using fresh drug every 12 h, and cells were grown under these conditions for
extracts of L8 muscle cells grown in differentiation medium for different 36 h (middle panel). In a control plate, L8 cells were grown only in the
time periods as indicated. Kinase activity of p38 was analyzed by presence of DM for 36 h (top panel). In a third plate, cells were treated
immunoprecipitation of p38. The immunocomplex was used to phospho- for 36 h with DM and SB 203580 and for an additional period of 24 h
rylate the substrate, GST-ATF2 protein, as described under “Experi- with DM only (bottom panel). Arrowheads point at multinucleated
mental Procedures.” Proteins were separated over SDS-PAGE, and the myotubes. Cells were photographed at 2003 magnification.
amount of phosphorylated GST-ATF2 was quantified using the Phos-
phorImager and plotted in the histogram. Maximal kinase activity was
set to 100 units. Average results from two independent experiments are of mitogen deprivation (Fig. 2) (28). However, the drug was
presented. Bars represent standard errors. B, the same protein extracts most probably not toxic to cells because the percentage of living
from L8 cells as in A were separated using SDS-PAGE, and phospho- cells was not changed in drug-treated culture, and these cells
rylated forms of p38 were identified by Western blotting with an anti-
recovered and differentiated following removal of the drug
body that recognized the dual phosphorylated form of p38 (upper
bands). The same blot was exposed also to an antibody that recognized (Figs. 2 and 3B).
all forms of p38 (lower bands). The control lanes (Cont.): 2, extract of C6 Activation of p38 with MKK6 Induces Muscle Differentiation
cells; 1, extract of anisomycine-treated C6 cells. The asterisk represents by MyoD—The dramatic effect of p38 inhibitor on the expres-
a nonspecific band. C, GST-MEF2C protein was phosphorylated in vitro sion of MyoD and MEF2 family members prompted us to in-
as described under “Experimental Procedures” using extracts of L8 cells
grown in differentiation medium for different time periods as indicated. vestigate whether p38 affected also the activity of the myogenic
In one case, L8 cells were grown for 48 h in DM in the presence of 20 mM regulator MyoD. The ectopic expression of MyoD converts
SB 203580. Isoforms of p38 were depleted from extracts in the following 10T1/2 fibroblasts to muscle (29). The effect of p38 on MyoD
way: antibodies to the different isoforms of p38 were added to extracts activity was studied by transiently transfecting 10T1/2 fibro-
followed by an immunoprecipitation procedure. p38-depleted extracts
were used to phosphorylate GST-MEF2C. Control lane, GST protein blasts with expression vectors of MyoD and wild type MKK6,
was used in a kinase assay with extract of L8 cells grown for 48 h in DM. the direct activator of p38. Most 10T1/2 fibroblasts transfected
with MyoD expression vector also expressed the differentiation
the expression of muscle specific genes like myogenin and marker MHC when grown for 24 h in differentiation medium
MLC2 was completely inhibited in cells treated with the drug (Fig. 4). However, if the transfected cells were continuously
(Fig. 3A). The expression of MEF2 that participates with the grown in high serum-containing medium (15% bovine calf se-
MyoD family in the differentiation process was also abolished rum), only 28% of the cells that expressed MyoD expressed also
in the drug-treated cells (Fig. 3A). Induction of the cyclin/ MHC (Fig. 4). Under these conditions, cells that were co-trans-
cyclin-dependent kinase inhibitor p21 (Waf-1) participating in fected with MKK6 exhibited a significantly higher proportion of
the exit of myoblasts from the cell cycle was inhibited in SB MHC staining in the cytoplasm of MyoD-expressing cells (69%
203580-treated cells. Consequently, the drug blocked differen- of the cells) (Fig. 4). On the other hand, addition of SB 203580
tiation completely. We noticed dead cells in both untreated and reversed the effect of MKK6 and completely inhibited MyoD,
treated cells grown in the serum-free medium (DM), which i.e. only about 5% of the cells that expressed MyoD also ex-
probably underwent programmed cell death under conditions pressed MHC. In a parallel experiment, we studied the effects
5196 Role of p38 Pathway in Myogenesis

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


FIG. 4. MyoD activity is up-regulated by MKK6 and down-
regulated by treatment of cells with SB 203580. A, 10T1/2 fibro-
blasts were transiently transfected either with MyoD expression vector
or with expression vectors of MyoD and MKK6. In one case, cells that
were transfected with MyoD and MKK6 were treated with 20 mM of SB
203580. Transfected cells were grown in the presence of high serum
(growth medium) for 24 h after which they were fixed and double-
stained for MHC (cytoplasmic staining) and for MyoD (nuclear stain-
ing). B, quantification of the results presented in A. Transfected cells
were grown as described in A, except in one case in which cells were
FIG. 3. SB 203580 inhibits the expression of muscle-specific grown in DM for 24 h. The percentage of differentiation was calculated
markers in L8 cells. A, L8 cells were grown in DM and in the presence by dividing the number of double-stained cells (MyoD and MHC) by the
or absence of SB 203580 for different periods of time as indicated. Total total number of MyoD-stained cells (single- and double-stained). Each
RNA was extracted from cells, separated over an agarose gel, and bar in the histogram represents the results of counting of about 100
blotted onto a filter. Specific transcripts were determined by hybridiz- transfected cells. C, the in vitro kinase activity of p38 was analyzed
ing the filter to different labeled DNA probes as indicated. Hybridiza- using extracts from transfected cells as described under “Experimental
tion to GAPDH was used to control for loading of RNA on the gel. B, L8 Procedures.”
cells were grown in DM in the presence or absence of SB 203580 as
indicated. In one plate, cells were treated for 48 h with DM and SB
mRNA levels of several muscle-specific genes were induced to
203580 and for an additional period of 48 h with DM only (lane 4). Total
RNA was extracted and processed as described for A. significant levels (Fig. 5, lane 3) (30). If cells were treated with
SB 203580 during that period, the induction of MEF2, myoge-
of MKK6 and SB 203580 on the in vitro kinase activity of p38 nin, and MLC2 expression was significantly inhibited, al-
in extracts from transfected 10T1/2 cells (Fig. 4C). As expected, though expression of chimeric MyoD-ER remained unchanged
MKK6 induced whereas SB 203580 inhibited p38 kinase activ- (Fig. 5, lanes 4 and 5). Therefore, the inhibitor either inacti-
ity. Like MKK6, ectopic expression of p38 isoforms, mainly vated the chimera MyoD protein or affected other transcription
p38g and d, in 10T1/2 cells strongly induced the ability of MyoD factors or co-activators that may function in concert with MyoD
to activate endogenous MHC (data not shown). Therefore we to initiate myogenesis.
conclude that activation of p38 MAPK pathway appears to MKK6 Augments Transcription of MCK via the MEF2-bind-
contradict the inhibitory effects of serum on the function of ing Site—To further analyze the effect of MKK6 on the tran-
MyoD as judged by the activation of endogenous MHC scriptional activity of MyoD, expression vectors of these pro-
expression. teins were co-transfected with a reporter gene that contained a
Another approach was used to investigate whether p38 af- minimal promoter and four MyoD-binding sites (4R-tk-CAT).
fected the activity of MyoD. We used a cell line expressing an Surprisingly, the activity of MyoD in the induction of this
estrogen receptor-MyoD chimera protein (21). The chimera pro- reporter gene was almost not affected by co-transfection of
tein remains inactive in the cytoplasm of cells. Treatment of MKK6 (Fig. 6A, lanes 9 –12). However, MyoD activity was
cells with estradiol induces the chimera protein that migrates increased more significantly by MKK6 in activating the tran-
to the nucleus and activates muscle-specific transcription. In scription of a reporter gene whose expression was driven by
this cell line, myogenesis is initiated by the activity of the MCK regulatory sequences (pe1AT-CAT) (Fig. 6A, lanes 1– 4).
chimera protein. Therefore, inhibition of MyoD in these cells is This reporter gene contained two MEF2-binding sites within
expected to abolish myogenesis. Activity of the chimera protein the MCK enhancer element that could contribute to the effect
was induced by the addition of estradiol to cells that were of MKK6 (12). Co-expression of MEF2C did not further poten-
grown in the presence or absence of SB 203580. Cells were tiate the transcriptional activity of MyoD on this promoter (not
grown in the presence of estradiol for 24 h, at which time shown). These results may be explained by previous results
Role of p38 Pathway in Myogenesis 5197

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


FIG. 5. Treatment with SB 203580 prevents muscle differenti-
ation of cells expressing a conditional MyoD-ER chimera pro-
tein. 10T1/2 cells that constitutively express a chimera protein of MyoD
and estrogen receptor hormone-binding domain were studied. MyoD
activity was induced by the addition of estradiol and differentiation
medium to cells. Some cells were treated with SB 203580 at different FIG. 6. p38 affects the transcription of MCK reporter genes via
concentrations added together with estradiol and DM, and RNA was the MEF2-binding sites. A, 10T1/2 fibroblasts were transiently trans-
extracted 24 h later. RNA was separated over agarose gel and analyzed fected with expression vectors of MyoD or MEF2C with or without the
by Northern blotting. Blot was repeatedly hybridized with labeled direct activator of p38, MKK6. Several reporter genes were used;
probes of MyoD, MEF2, myogenin, MLC2, and GAPDH, which was used pe1AT-CAT carried a regulatory sequence of MCK enhancer that con-
to control for loading of RNA. RNA was underloaded in lane 2 as can be tained two MEF2-binding sites and MCK promoter; p110 MCK CAT
noticed by the appearance of a weak GAPDH band. carried a smaller MCK enhancer that contained one MEF2-binding site
and MCK promoter; 4R-tk-CAT that contained four MyoD-binding sites
that indicated that endogenous MEF2 expression was stimu- in the promoter region and MEF2-MCK-CAT that contained three
MEF2-binding sites and MCK promoter. All reporter genes contained
lated by the expression of myogenin in fibroblast cells (31). For the CAT reading frame. Protein extracts of the transfected cells were
this reason, we did not co-transfect a MEF2C expression vector used in a CAT assay according to the transfection efficiency that was
in the subsequent experiments that analyzed MCK regulatory measured as described under “Experimental Procedures.” The chloram-
sequences. To study the possible role of the MEF2 sites in the phenicol products were separated by thin layer chromatography and
quantified by phosphor imaging (Fuji). For each reporter gene, the
stimulation of MyoD activity by MKK6, a similar transfection maximal CAT activity was set to 100 units. Average results from three
experiment was done with an MCK reporter gene that con- independent experiments are presented. Error bars represent standard
tained the MyoD-binding sites and only one MEF2 site (p110- errors. B, the same constructs described in A were used to transfect
MCK-CAT) (17). Induction of this reporter gene by MyoD was 10T1/2 cells with the difference that some plates were treated immedi-
ately after transfection with 10 mM SB 203580. Also, the expression
only mildly affected by the co-expression of MKK6 (Fig. 6A,
vector of GAL4-VP16 was co-transfected with the pGAL-CAT reporter
lanes 5– 8). To prove MKK6 activated MCK via the MEF2 site, gene. For each reporter gene, the maximal CAT activity was set to 100
we generated a reporter gene that contained multiple MEF2 units. Average results from three independent experiments are pre-
sites of the MCK enhancer and a basal promoter of MCK sented. Error bars represent standard errors.
(MEF2-MCK-CAT). Indeed, this promoter was induced by the
expression of MEF2C and was further induced by MKK6 (Fig. MCK reporter gene via the MEF2-binding sites.
6A, lanes 13–15). Expression of MKK6 alone was sufficient to A Transcriptionally Inactive MEF2C Protein Abrogates the
induce this promoter in a significant fashion (Fig. 6A, lane 16). Effects of MKK6 on the MCK Enhancer—To study the role of
We conclude that MEF2 sites probably play a role in the stim- MEF2 in mediating the effects of p38 on the transcription of the
ulation of MCK transcription observed in the presence of MCK reporter gene, we generated a transcriptionally inactive
MKK6. Addition of the p38 inhibitor, SB 203580, significantly MEF2C protein (see “Experimental Procedures”). This protein
blocked the transcriptional activity of MyoD in the induction of did not contain a large part of its transactivation domain that
the two MCK enhancer reporter genes, (pe1AT-CAT and p110- included two phosphorylation sites of p38 (MEF2C-D) (12). A
MCK-CAT) (Fig. 6B, lanes 1– 4), and more modestly affected its similar MEF2A protein that functioned as a dominant negative
activity on the minimal promoter driven by E boxes (4R-tk- was recently described by Ornatsky and colleagues (32). By
CAT) (Fig. 6B, lanes 5 and 6). The transcriptional activity of itself, MEF2C-D retained minimal transcriptional activity com-
MEF2 on a basal promoter driven by MEF2 sites was blocked pared with the wild type protein (Fig. 7A, compare lanes 2 and
by SB203580 (Fig. 6B, lanes 9 and 10). All in all, these results 6). When co-expressed with wild type MEF2C, the mutant
suggest that p38 modulates the transcriptional activity of the protein was able to repress the transcriptional activity of the
5198 Role of p38 Pathway in Myogenesis

FIG. 8. Transcriptional activity of GAL4-MEF2C protein is in-


duced, whereas activities of GAL4-MyoD proteins are not af-

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


fected by MKK6. 10T1/2 fibroblasts were transfected with different
expression constructs as indicated and the pGAL4-CAT reporter gene.
Cells were lyzed 48 h after transfection, and protein extracts of the
transfected cells were used in a CAT assay according to the transfection
efficiency that was measured as described under “Experimental Proce-
dures.” The chloramphenicol products were separated by thin layer
chromatography and quantified by phosphor imaging (Fuji). Average
results from three independent experiments are presented. Error bars
represent standard errors.
FIG. 7. A transcriptionally inactive MEF2C protein abrogates
the effects of MKK6 on the MCK enhancer. A, MEF2C-D represses The p38 Pathway Affects the MEF2C Protein but Not the
the transcriptional activity of wild type MEF2C protein. 10T1/2 fibro- MyoD Protein—Although the results presented in Fig. 6 sug-
blasts were transfected with different expression vectors and reporter
genes as indicated. Reporter genes: Pe1AT-CAT contains enhancer and gest that the p38 pathway operates via MEF2 sites, we could
promoter elements of MCK, 4R-tk-CAT contains four MyoD-binding still notice an effect on MyoD (Fig. 6, A, lanes 9 –12, and B,
sites and the basal tk promoter, and pGAL-CAT contains five GAL4- lanes 5 and 6). This effect on MyoD may result from its asso-
binding sites and a basal tk promoter. MEF2C-D expression vector was ciation with MEF2, which is known to function as a co-activator
transfected at 2 mg (31), 4 mg (32), and 8 mg (34). Cells were lyzed 48 h
after transfection, and protein extracts of the transfected cells were of MyoD (18). To study the effect of p38 on MyoD independently
used in a CAT assay according to the transfection efficiency that was of MEF2, we used the GAL4 activator/reporter system. The
measured as described under “Experimental Procedures.” The chloram- transcription activators in this system were composed of MyoD
phenicol products were separated by thin layer chromatography and fragments that were fused to the DNA-binding domain of yeast
quantified by phosphor imaging (Fuji). For each reporter gene, the
maximal CAT activity was set to 100 units. Average results from two
transcription factor GAL4. The chimeric activators are ex-
independent experiments are presented. B, MEF2C-D represses the pected to bind to the GAL4 DNA-binding sites of a reporter
induction of MCK transcription mediated by MyoD and MKK6. 10T1/2 gene and activate transcription via the transactivation domain
fibroblasts were transfected with different plasmid DNA as indicated. of MyoD. MEF2 was suggested to interact with the DNA-
Reporter gene and expression vectors are described in A. MEF2C-D
binding domain of MyoD-E12 heterodimers (18). Two MyoD
expression vector was transfected at 2 mg (31) and 4 mg (32). Trans-
fection, CAT assay and processing of the results were done as described proteins that were not expected to interact with MEF2 were
for A. Average results from three independent experiments are pre- used; one that did not contain the HLH domain (GAL-DHLH
sented. Error bars represent standard errors. MyoD) and another that contained a substituted DNA-binding
domain from the Drosophila acheate-scute protein (GAL-
former protein (Fig. 7A, lanes 2–5). However, the mutant T4basic MyoD). The transcription mediated by GAL4-MyoD
MEF2C protein did not repress activation by MyoD (lanes 8 proteins was not affected by co-expression of MKK6 (Fig. 8,
and 9) or GAL4-VP16 (lanes 11 and 12) and therefore was lanes 7–12). However, MKK6 augmented the activity of a chi-
specific to wild type MEF2C. To find out whether the mutant meric GAL4-MEF2C transcription factor (Fig. 8, lanes 1 and 2).
protein could block the effect of p38 on the MCK regulatory The activity of two MEF2C mutants that did not contain their
sequences, we transfected the different expression plasmids p38 phosphorylation sites (293, 300A and 387A) was not sig-
with the MCK reporter gene (pe1AT-CAT). The transcription- nificantly increased by the expression of MKK6 (Fig. 8, lanes
ally inactive MEF2C protein inhibited the induced expression 3– 6). Therefore, we conclude that the transcriptional activity of
of MCK reporter mediated by MyoD (Fig. 7B, lanes 1– 4). More- GAL4-MyoD was not affected, whereas the activity of GAL4-
over, it also abolished the additional activity contributed by MEF2C was affected by MKK6.
MKK6 (Fig. 7B, lanes 5–7). The inhibition was specific to the MEF2C Is Phosphorylated by p38 in Muscle Cells—To dem-
MCK enhancer that contained the MEF2-binding site, because onstrate that GAL4-MEF2C protein is phosphorylated in cells
the inactive MEF2C protein did not inhibit MyoD activation of by p38, 293 cells were transfected with expression vector of
reporter gene containing only MyoD-binding sites (4R-tk-CAT- GAL4-MEF2C alone or GAL4-MEF2C with MKK6 (Fig. 9A). In
)(Fig. 7A, lanes 8 and 9). We conclude that MKK6 induced the a duplicate set of transfected plates, the cells were treated with
transcription of MCK via MEF2 protein(s) and that this induc- SB 203580. Proteins were metabolically labeled with [35S]me-
tion was blocked by competition of the transcriptionally inac- thionine, and GAL4-MEF2C was immunoprecipitated. The mo-
tive MEF2C protein. bility of the GAL4-MEF2C protein in the gel indicated its
Role of p38 Pathway in Myogenesis 5199
ited by treatment of cells with SB 203580 (Fig. 9B, lane 8).
Consequently, MEF2 proteins are phosphorylated by p38 in L8
muscle cells.
DISCUSSION
In this report we suggest for the first time the possible
involvement of p38 in skeletal muscle differentiation. We show
that the activity of p38 was induced in L8 myoblasts during the
formation of multinucleated myotubes. Interference with p38
activity by the specific inhibitor SB 203580 completely abol-
ished muscle fusion and expression of all myogenic markers
that were tested. We studied the effects of p38 MAPK on
muscle-specific transcription and found that its activator,
MKK6, stimulated the expression of muscle-specific genes, and
the effect was mediated by MEF2C. The latter was demon-
strated before to be an essential transcription activator in
myogenesis (13, 33, 34).
Members of the MEF2 and MyoD families act within a reg-
ulatory network that establishes the differentiated phenotype
of skeletal muscle (13). MyoD and MEF2 family members work
in concert to activate the expression of many muscle-specific
genes including their own. Therefore, inhibition of the activity

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


of either MEF2 or MyoD family members results in complete
inhibition of skeletal muscle differentiation. p38 MAPK is a
potential activator of MEF2C and as part of the MEF2-MyoD
circuit it is expected to control muscle differentiation.
That MEF2 family members played a part in mediating p38
function in the activation of MCK transcription and myogen-
esis was suggested by the following. 1) Induction of MCK re-
porter gene by MyoD was augmented by MKK6 only if MEF2
sites were present at the regulatory sequences. MKK6 did not
significantly affect the activation of MyoD from promoters that
FIG. 9. Phosphorylation of MEF2 proteins in cells. A, 293 cells did not contain MEF2 site (Fig. 6). 2) The effect of MKK6 was
transfected with GAL4 (lanes 8 and 9) or GAL4-MEF2C (lanes 1–7) specifically abolished by the expression of a transcriptionally
expression vector alone or with MKK6 were labeled with [35S]methi- inactive MEF2C protein (Fig. 7). 3) The transcriptional activity
onine. As indicated, some transfected cells were treated with SB 203580
(20 mM) for 36 h before proteins were extracted. GAL4 proteins were
of GAL4-MEF2C fusion proteins was induced, whereas the
immunoprecipitated from protein extracts and separated using SDS- activity of GAL4-MyoD proteins was not changed by MKK6
PAGE. Lanes 5–7, immunoprecipitates were treated with alkaline phos- (Fig. 8).
phatase. The asterisks represent truncated forms of GAL4-MEF2C. B, MEF2C is not the only member of the family that is ex-
dividing myoblasts (0 h in DM) and differentiating myotubes (48 h in
pressed in muscle. In fact MEF2A protein accumulates before
DM) were metabolically labeled with [35S]methionine (lanes 1– 4) or
with [32P]orthophosphate (lanes 5–9). MEF2A and MEF2C proteins MEF2C, which is expressed later during the differentiation of
were immunoprecipitated and separated over SDS-PAGE. In lanes 3 myoblasts (35, 36). For that reason we should expect that other
and 7, a competitive peptide against which the MEF2 antibody was members of the family may be similarly regulated by p38.
made was added to extracts prior to the immunoprecipitation proce- Interestingly, MEF2A contains a serine at the position equiv-
dure. Lanes 4 and 9 are control lanes; MEF2 antibody was not added to
the immunoprecipitation reaction. alent to Ser387 found in MEF2C (37), suggesting that it may be
regulated similarly. Indeed, recent studies suggest that
phosphorylation status. MEF2C was phosphorylated by p38 in MEF2A is phosphorylated by p38.2
cells because it migrated faster in cells treated with SB 203580 We noticed that inhibition of p38 in muscle cells also abol-
(Fig. 9A, compare lanes 1 and 2). Ectopic expression of MKK6 ished the expression of MEF2 (Figs. 3A and 5). Inhibition of
in the transfected cells resulted in a smear of slower migrating MEF2 expression may be a result of the repression of MEF2C
GAL-MEF2C species (Fig. 9A, lane 3). Phosphorylation of that functions to induce its own transcription and/or the re-
MEF2C was confirmed by treatment of the immunoprecipitates pression of other transcription factors such as MyoD or Myf5.
with alkaline phosphatase that compressed the smear to a Our results do not rule out the possibility that MyoD was
tight faster migrating band (Fig. 9A, lanes 5–7). Unlike GAL4- directly affected by p38, because the reporter gene regulated by
MEF2C, the migration of transfected GAL4 protein was not MyoD only, 4R-tk-CAT, was mildly affected by MKK6 or SB
affected by the co-expression of MKK6 (Fig. 9A, lanes 8 and 9). 203580 (Fig. 6). Other transcription factors activated by p38,
Therefore, both the expression of MKK6 and treatment with like ATF2, CREB, and Elk-1 may participate in the differenti-
SB 203580 affected the phosphorylation state of transfected ation process induced by p38.
GAL4-MEF2C protein in cells. Four isoforms of p38 are known (a, b, g, and d). We suggest
To find out if MEF2 proteins are phosphorylated in muscle that p38a and b are the major isoforms involved in differenti-
cells, we immunoprecipitated endogenous MEF2 proteins from ation of L8 cells because 1) The inhibitor, SB 203580, that is
L8 cells (Fig. 9B). MEF2 proteins were detected mainly in specific to the a and b isoforms (38 – 40) inhibited the differen-
differentiated myotubes (Fig. 9B, lanes 1 and 2). Differentiated tiation of L8 cells (Figs. 2 and 3), and 2) immunodepletion of the
cells were phosphate-labeled, and MEF2 proteins were isolated a and b but not of g and d isoforms from L8 extracts signifi-
to learn whether these proteins were phosphorylated in myo- cantly inhibited the phosphorylation of GST-MEF2C (Fig. 1C).
tubes. MEF2 proteins are phosphorylated in muscle cells (Fig.
2
9B, lane 6). Phosphorylation of MEF2 proteins is partly inhib- J. McDermott, personal communication.
5200 Role of p38 Pathway in Myogenesis
Interestingly, our preliminary studies suggest that g and d 201–216
5. Moxham, C. M., Tabrizchi, A., Davis, R. J., and Malbon, C. C. (1996) J. Biol.
isoforms are more efficient than a and b isoforms in the induc- Chem. 271, 30765–30773
tion of muscle differentiation when transfected with MyoD. 6. Tsakiridis, T., Taha, C., Grinstein, S., and Klip, A. (1996) J. Biol. Chem. 271,
19664 –19667
Farther studies will elucidate the role of p38 isoforms in muscle 7. Rouse, J., Cohen, P., Trigon, S., Morange, M., Alonso-Liamazares, A.,
differentiation. Zamanillo, D., Hunt, T., and Nebrwda, A. R. (1994) Cell 78, 1027–1037
Recently, we reported that ERK MAPK activity was induced 8. Raingeaud, J., Gupta, S., Rojers, J. S., Dickens, M., Han, J., Ulevitch, R. J., and
Davis, R. J. (1995) J. Biol. Chem. 270, 7420 –7426
during the differentiation of C2 cells and that this activity 9. Derijard, B., Raingeaud, J., Barrett, T., Wu, I.-H., Han, J., Ulevitch, R. J., and
promoted muscle differentiation (25). The similarities in the Davis, R. J. (1995) Science 267, 682– 685
pattern of activities of p38 and ERK in muscle cells imply that 10. Han, J., Lee, J.-D., Jiang, Y., Li, Z., Feng, L., and Ulevitch, R. J. (1996) J. Biol.
Chem. 271, 2886 –2891
these two distinct pathways have similar effects on muscle 11. Taha, C., Tsakiridis, T., McCall, A., and Klip, A. (1997) Am. J. Physiol. 273,
differentiation. However, our data suggest that the two path- 68 –76
12. Han, J., Jiang, Y., Li, Z., Kravchenko, V. V., and Ulevitch, R. J. (1997) Nature
ways also exhibit distinct activities. Firstly, inhibition of ERK 386, 296 –299
with PD 098059 only partially prevented the fusion of myo- 13. Olson, E. N., Perry, M., and Schulz, R. A. (1995) Dev. Biol. 172, 2–14
blasts and did not inhibit the expression of muscle-specific 14. Zechner, D., Thuerauf, D. J., Hanford, D. S., McDonough, P. M., and
Glembotski, C. C. (1997) J. Cell Biol. 139, 115–127
markers, whereas inhibition of p38 with SB 203580 prevented 15. Tapscott, S. J., Davis, R. L., Thayer, M. J., Cheng, P. F., Weintraub, H., and
fusion of myoblasts and expression of muscle-specific markers. Lassar, A. B. (1988) Science 242, 405– 411
Secondly, the activities of ERK and p38 are differentially in- 16. Weintraub, H., Davis, R., Lockshon, D., and Lassar, A. (1990) Proc. Natl. Acad.
Sci. U. S. A. 87, 5623–2627
duced; p38 is induced earlier than ERK in cells grown in 17. Buskin, J. N., and Hauschka, S. D. (1989) Mol. Cell. Biol. 9, 2627–2640
differentiation medium (Ref. 25 and present work). These dif- 18. Molkentin, J. D., Black., B. L., Martin, J. F., and Olson, E. N. (1995) Cell 83,
1125–1136
ferences suggest that the two pathways perform distinct func- 19. Weintraub, H., Dwarki, V. J., Verma, I., Davis, R., Hollenberg, S., Snider, L.,
tions and that their combined activity may be required for the Lassar, A., and Tapscott, S. J. (1991) Genes Dev. 5, 1377–1386
differentiation process. 20. Yaffe, D., and Saxel, O. (1977) Nature 270, 725–727

Downloaded from http://www.jbc.org/ by guest on November 24, 2014


21. Hollenberg, S. M., Cheng, P. F., and Weintraub, H. (1993) Proc. Natl. Acad.
Studies of different cell lineages suggest that a distinct bal- Sci. U. S. A. 90, 8028 – 8032
ance between the different MAPK pathways is essential for the 22. Davis, R. L., Cheng, P.-F., Lassar, A. B., and Weintraub, H. (1990) Cell 60,
survival of these cells. During maturation of HL-60 cells to the 733–746
23. An, G., Hikada, K., and Siminovitch, L. (1982) Mol. Cell. Biol. 2, 1628 –1632
neutrophil phenotype, the activity of JNK was reduced, 24. Gorman, C. M., Moffat, L. F., and Howard, B. H. (1982) Mol. Cell. Biol. 2,
whereas the activity of p38 was induced (41). In rat pheochro- 1044 –1051
25. Gredinger, E., Gerber, A. N., Tamir, Y., Tapscott, S. J., and Bengal, E. (1998)
mocytoma PC-12 cells that serve as a model for neuronal dif- J. Biol. Chem. 273, 10436 –10444
ferentiation, induced activity of ERK MAPK and reduced ac- 26. Hibi, M., Lin, A., Smeal, T., Minden, A., and Karin, M. (1993) Genes Dev. 7,
tivities of JNK and p38 pathways were critical for the survival 2135–2148
27. Cuenda, A., Rouse, J., Doza, Y. N., Meier, R., Cohen, P., Gallagher, T. F.,
of these cells (42). The induction of p38 in myocardial cells Young, P. R., and Lee, J. C. (1995) FEBS Lett. 364, 229 –233
served to protect them from apoptosis in one study (43) or to 28. Wang, J., and Walsh, K. (1996) Science 273, 359 –361
29. Davis, R. L., Weintraub, H., and Lassar, A. B. (1987) Cell 51, 987–1000
increase apoptosis in another study (44). In these cellular mod-
30. Gerber, A. N., Klesert, T. R., Bergstrom, D. A., and Tapscott, S. J. (1997) Genes
els, cell survival or programmed cell death happens as a result Dev. 11, 436 – 450
of a balance between the activities of ERK, JNK, and p38 31. Cserjesi, P., and Olson, E. N. (1991) Mol. Cell. Biol. 11, 4854 – 4862
32. Ornatsky., O. I., Andreucci, J. J., and McDermott, J. C. (1997) J. Biol. Chem.
MAPKs. Therefore, it is possible that the induction of ERK and 272, 33271–33278
p38 MAPKs during skeletal muscle differentiation plays a role 33. Edmonson, D. G., Cheng, T., Cserjesi, P., Chakraborty, T., and Olson, E. N.
in preventing programmed cell death as well as in the activa- (1992) Mol. Cell. Biol. 12, 3665–3677
34. Lilly, B., Zhao, B., Ranganayakulu, G., Paterson, B. M., Schulz, R. A., and
tion of muscle-specific genes. Olson, E. N. (1995) Science 267, 688 – 693
35. Martin, J. F., Schwarz, J. J., and Olson, E. N. (1993) Proc. Natl. Acad. Sci.
Acknowledgments—We thank Dr. Uri Nudel and Dr. David Yaffe for U. S. A. 90, 5282–5286
L8 cells. We thank Dr. Stephen J. Tapscott for the 10T1/2 MyoD-ER cell 36. McDermott, J. C., Cardoso, M. C., Yu, Y.-T., Andres, V., Leifer, D., Krainc, D.,
line and muscle reporter genes. We thank Dr. Jiahuai Han for MKK6 Lipson, S. A., and Nadal-Ginard, B. (1993) Mol. Cell. Biol. 13, 2564 –2577
and GAL4-MEF2C constructs and the antibodies to different isoforms of 37. Kato, Y., Kravchenko, V. V., Tapping, R. I., Han J., Ulevitch, R. J., and Lee,
p38. We thank Dr. Ami Aronheim and Dr. Michael Karin for offering us J.-D. (1997) EMBO J. 23, 7054 –7066
38. Jiang, Y., Gram, H., Zhao, M., New, L., Gu, J., Feng, L., Di Padova, F.,
the pGEX-ATF2 plasmid. We thank Dr. Eric Olson for the MEF2C Ulevitch, R. J., and Han, J. (1997) J. Biol. Chem. 272, 30122–30128
expression vector. We thank Dr. Bianca Raikhlin-Eisenkraft for critical 39. Cuenda, A., Cohen, P., Buee-Scherre, V., and Goedert, M. (1997) EMBO J. 16,
reading of the manuscript. 295–305
40. Enslen, H., Raingeaud, J., and Davis, R. J. (1998) J. Biol. Chem. 273,
REFERENCES 1741–1748
1. Weintraub, H., Davis, R., Tapscott, S., Thayer, M., Krause, M., Benezra, R., 41. Frasch, S. C., Nick, J. A., Fadok, V. A., Bratton, D. L., Worthern, G. S., and
Blackwell, T. K., Turner, D., Rupp, R., Hollenberg, S., Zhuang, Y., and Henson, P. M. (1998) J. Biol. Chem. 273, 8389 – 8397
Lassar, A. (1991) Science 251, 761–766 42. Xia, Z., Dickens, M., Raingeaud, J., Davis, R. J., and Greenberg, M. E. (1995)
2. Lassar, A. B., Skapek, S. X., and Novitch, B. (1994) Curr. Opin. Cell Biol. 6, Science 270, 1326 –1331
788 –794 43. Zechner, D., Craig, R., Hanford, D. S., McDonough, P. M., Sabbadini, R. A., and
3. Coleman, M. E., DeMayo, F., Yin, K. C., Lee, H. M., Geske, R., Montgomery, C., Glembotski, C. C. (1998) J. Biol. Chem. 273, 8232– 8239
and Schwartz, R. J. (1995) J. Biol. Chem. 270, 12109 –12116 44. Wang, Y., Huang, S., Sah, V. P., Ross, J., Jr., Brown, J. H., Han, J., and Chien,
4. Florini, J. R., Ewton, D. Z., and Magri, K. A. (1991) Annu. Rev. Physiol. 53, K. R. (1998) J. Biol. Chem. 273, 2161–2168

You might also like