Вы находитесь на странице: 1из 11

IUBMB Life, 58(11): 621 631, November 2006

Critical Review

ERK1/2 MAP Kinases in Cell Survival and Apoptosis


Zhimin Lu1 and Shuichan Xu2
1
Brain Tumor Center and Departments of Neuro-Oncology and Molecular Genetics, The University of Texas M. D.
Anderson Cancer Center, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas,
USA
2
Signal Pharmaceuticals, LLC, 4550 Towne Centre Ct., San Diego, California, USA

ERK 3 to 8 have been identied, although their function and


Summary regulation are less well characterized (7). The MAP kinase
ERK1/2 is an important subfamily of mitogen-activated protein signaling pathway is a three-tiered cascade: a MAP kinase is
kinases that control a broad range of cellular activities and activated by a MAP kinase kinase (MKK or MAP2K), which
physiological processes. ERK1/2 can be activated transiently or is in turn activated by a MAP kinase/ERK kinase (MEK)
persistently by MEK1/2 and upstream MAP3Ks in conjunction with kinase or MAP kinase kinase kinase (MEKK or MAP3K)
regulation and involvement of scaolding proteins and phospha- (8, 9) (Fig. 1). In mammalian cells, 12 MAP kinases, 7
tases. Activation of ERK1/2 generally promotes cell survival; but
under certain conditions, ERK1/2 can have pro-apoptotic functions. MAP2Ks, and 20 MAP3Ks have been identied. MAP kinases
IUBMB Life, 58: 621631, 2006 are activated by dual phosphorylation on a conserved Thr-
Xaa-Tyr motif in their activation loop by an upstream
MAP2K. MAP2Ks are themselves activated by phosphoryla-
Keywords ERK1/2; cell survival; apoptosis.
tion via an upstream MAP3K (10). In addition to the
predominant mode of MAP kinase activation by MAP2K-
INTRODUCTION: MAP KINASE PATHWAYS AND THEIR mediated dual phosphorylation, MAP2K-independent activa-
SPECIFICITY tion of p38 MAP kinase has been reported. Transforming
With a total of 518 members identied in the human genome, growth factor b-activated protein kinase 1 (TAK1)-binding
protein kinases are one of the largest gene families in eukaryotes protein 1 (TAB1), a scaolding protein, binds to p38a-MAP
(1 3). Mutation and dysregulation of protein kinases play kinase and stimulates autophosphorylation of its Thr-Gly-Tyr
causal roles in human disease and provide a basis for developing motif, resulting in the initiation of kinase activity indepen-
agonists and antagonists of these enzymes for therapy. Acting as dently of MKK3 and MKK6 (11).
serine and threonine protein kinases, mitogen-activated protein Each of the MAP kinase families can be activated by at
(MAP) kinases comprise a family of protein kinases whose least two cognate MAP2Ks and by multiple MAP3Ks.
function and regulation have been conserved evolutionarily, MAP2Ks display remarkable substrate specicity for their
from unicellular organisms like yeast to complex organisms MAP kinases; several recognize only one or two MAP kinases
such as humans (4). By phosphorylating specic serines and and few if any other substrates. In contrast, MAP3Ks can
threonines of target protein substrates, MAP kinases regulate a activate multiple MAP kinase cascades (10) (Fig. 1). The
wide range of processes: cell growth and dierentiation, gene multiple mammalian MAP3Ks, MAP2Ks, and MAPKs form
expression, mitosis, cell motility, metabolism, cell survival and distinct signal transduction pathways and complexes of signal-
apoptosis, and embryogenesis (5, 6). ing molecules in response to dierent extracellular stimuli. The
The classic MAP kinase family consists of three sub- dierent combinations of these three tiers of kinases give rise to
families: extracellular signal-regulated kinase (ERK; ERK1 the signaling specicity. For example, the MAP3K TAK1 is
and ERK2), c-Jun N-terminal kinase (JNK; JNK1, JNK2, indispensable for activation of JNK in response to the pro-
and JNK3), and p38-MAP kinase (a, b, d, and g). In addition, inammatory molecule lipopolysaccharide (LPS), whereas the
expression of four MAP3Ks is required for osmotic stress-
Received 27 April 2006; accepted 14 August 2006
Address correspondence to: Zhimin Lu, The University of Texas induced JNK activation (12). The signaling specicity is also
M. D. Anderson Cancer Center, Houston, TX 77030, USA. controlled by regulation of scaolding proteins, which can
E-mail: zhiminlu@mdanderson.org sequester and insulate signaling components and direct them to
ISSN 1521-6543 print/ISSN 1521-6551 online 2006 IUBMB
DOI: 10.1080/15216540600957438
622 LU AND XU

specic subcellular localizations, enhancing the signal ux known as FXF) binding motif, which consists of two Phe
and mediating cross-talk with other pathways (13). residues separated by one residue and is often followed by a
MAP kinases directly interact with scaolding proteins, Pro residue (FXFP) (18 20). This docking motif binds to a
activators, and eectors, and these interactions also govern hydrophobic pocket in the large lobe of the MAP kinase
signaling specicity. MAP kinases have a docking groove adjacent to its catalytic domain. The DEF motif is within 10
comprised of an acidic common docking (CD) domain and residues COOH-terminal to the site of phosphorylation on the
Glu-Asp (ED) pockets that are distinct from their active sites substrate, which corresponds to the distance between the
(14, 15). The interacting proteins, such as MAP kinase hydrophobic region and the catalytic sites in ERK2 (21).
substrates, MAP2Ks, phosphatases, and scaolds, have a Unlike the D domain of substrates that can bind to active and
docking (D) domain (DEJL motif-docking site for ERK and inactive MAP kinase, the DEF motif only binds to active
JNK, also known as Leu-X-Leu motif) that complements the ERK2 molecules, because the DEF binding site on ERK is
MAP kinase binding motifs. The D domain is composed of exposed only upon ERK2 phosphorylation (21, 22). Thus, the
basic and hydrophobic residues with the consensus sequence substrate specicity and binding anity are controlled by the
Arg/Lys2-Xaa2 6-Faa-Xaa-Faa (where Faa represents the interaction between the phosphoacceptor motif, the D
hydrophobic residues Leu, Ile, and possibly Val) (14, 16, 17). domain, and the DEF motif in substrates and the correspond-
In addition to the D domain, many substrates of ERK and ing catalytic area, docking groove, and DEF binding region on
p38aMAP kinase have the DEF (docking site for ERK, also a MAP kinase (Fig. 2). In summary, the functional

Figure 1. Conventional (ERK1/2, JNK, and p38-MAP kinase) and ERK5 MAP kinase signaling pathways in mammalian cells
and in yeast. Only representative signaling molecules are shown. Solid arrows indicate main pathways, and dashed arrows
indicate branched pathways and indirect activation. MAPK, MAP kinase.

Figure 2. Representative schematic interactions between ERK2 and ERK2 substrates such as Elk1. The phosphoacceptor motif,
the D domain, and the DEF motif in ERK substrates interact with the corresponding catalytic area, docking groove, and DEF
binding region in ERK2.
ROLE OF ERK1/2 MAP KINASES IN CELLS 623

consequence of MAP kinase signaling activation is determined which indicates that ERK1 has a specic role in thymocyte
by dierent combinations of the three tiers of kinases; development (36). ERK1-decient mice exhibit a dramatic
regulation and involvement of distinct scaolding proteins; enhancement in striatum-dependent long-term memory, cor-
and selective interaction between the MAP kinase and its relating with facilitation of long-term potentiation in the
binding proteins, such as MAP kinase substrates, MAP2Ks, nucleus accumbens (37). Stimulus-dependent ERK2 signaling
phosphatases, and scaolding proteins. is increased in ERK17/7 mice suggesting that upregulation
of ERK2 occurs in the absence of ERK1 (37). In contrast to
the dispensable role of ERK1 in embryonic development that
ANTI-APOPTOTIC AND PRO-APOPTOTIC FUNCTIONS
may be due to ERK2 compensating for a deciency in ERK1,
OF ERK1/2 MAP KINASES
ERK2-decient embryos fail to form mesoderm (38), the
Overview of ERK1/2 ectoplacental cone, and extra-embryonic ectoderm giving rise
ERK1 and ERK2 are 83% identical with most of the to the fetal part of the placenta and the secondary trophoblast
dierences falling outside the kinase core (23). ERK1/2 is giant cells (39, 40). A severe defect in the development of the
expressed in all tissues. In broblasts, it is activated by growth placenta is likely an important cause of embryonic lethality
factors, serum, ligands that stimulate heterotrimeric G (39). Thus, ERK1 and ERK2 each have distinct roles in
protein-coupled receptors, cytokines, transforming growth physiological and developmental processes.
factors, osmotic stress, and microtubule disorganization (5).
Conventionally, ERK1/2 is activated by a cascade comprised Anti-apoptotic Functions of ERK1/2
of a small G protein Ras-Raf family member (Raf-1, A-Raf, Activation of ERK1/2 has been shown to inhibit apoptosis
B-Raf) followed by MEK1/2. Nevertheless, Ras is not the only in response to a wide range of stimuli, such as tumor necrosis
activator upstream of Raf for regulating ERK1/2. Protein factor (TNF), Fas ligand, TNF-related apoptosis-inducing
kinase C (PKC) a has been shown to phosphorylate and ligand (TRAIL) (41), radiation (42, 43), osmotic stress (33, 44,
activate Raf-1 (13). Mixed lineage kinase 3 (MLK3) is 45), hypoxia (46), growth factor withdrawal (47), nitric oxide
required for mitogen-stimulated phosphorylation of B-Raf at (48), hydrogen peroxide (49), matrix detachment (50), and
Thr598 and Ser601, and this phosphorylation is required for chemotherapeutic agents (51). In response to most of these
B-Raf activation and subsequently ERK1/2 activation (24). stimuli, ERK1/2 undergoes either transient or prolonged
Similarly, Raf is not the only MAP3K for regulating ERK1/2. activation, resulting in an anti-apoptotic eect. In contrast,
Mos (25), TPL2 protooncogene (26), MLK-like mitogen- inhibition of ERK1/2 promotes apoptosis. The anti-apoptotic
activated protein triple kinase (MLTK) (27), and interleukin-1 eect can be diminished after prolonged stimulation, because
(IL-1) receptor-associated kinase (IRAK) (28) have been ERK1/2 activity can be downregulated by dephosphorylation
reported to function as MAP3Ks to activate ERK1/2. or degradation of ERK1/2. In response to osmotic stress,
Overexpression of MEKK1, MEKK2, or MEKK3, which ERK1/2 is activated and increases its binding to MEKK1
are MAP3Ks that primarily regulate the JNK pathway, E3 ligase through the CD domain. Disruption of this
activates ERK1/2 indirectly through MEK1/2 (29 33). Thus, binding by mutating the ERK2 CD domain sustains ERK2
ERK1/2 can be activated by distinct MAP3Ks and MAP3K activation by reduced ubiquitination and degradation of
upstream activators in response to dierent stimuli. ERK2. Ubiquitination-resistant ERK2 with a mutant CD
ERK1/2 preferentially phosphorylates substrates that con- domain has an anti-apoptotic eect against osmotic stress-
tain the consensus sequence Pro-Xaa-Ser/Thr-Pro, whereby induced apoptosis (33). Thus, prevention of downregulation of
the Pro residue is conserved at the P1 position and the Pro ERK1/2 activity can promote cell survival.
residue at the P-2 position is less conserved (34). A Pro residue The anti-apoptotic eect of ERK1/2 can be regulated
at position P-1 can reduce the eciency of phosphorylation indirectly by p38 signaling (52 54). Inhibition of p38 MAPK
(35). The activation of ERK1/2, induced by dierent initiating stimulates Raf and ERK activities and induces myoblast
signals, results in the phosphorylation of dierent substrates; proliferation (52). Activation of p38 results in rapid depho-
thus far, more than 150 substrates have been identied. These sphorylation of MEK1/2 and subsequent apoptosis in human
substrates can be categorized as transcription factors, protein skin broblasts, whereas inhibition of p38 activity potently
kinases, protein phosphatases, cytoskeletal proteins, scaold- inhibits MEK1/2 dephosphorylation and apoptosis (53).
ing proteins, receptors, signaling molecules, apoptosis-related Furthermore, PP2A has been found in association with
proteins, as well as other types of proteins (Table 1) (22). MEK1/2 and ERK1/2 in cardiac ventricular myocytes.
The roles of ERK1 and ERK2 in embryogenesis have been Inhibition of p38 MAPK blocked the ERK-associated PP2A
illustrated by the study of ERK1/2 decient mice. ERK17/7 activity accompanied with enhanced ERK1/2 activity and
mice are viable, fertile, and of normal size (36). However, attenuated apoptosis in response to H2O2 stimulation (54).
thymocyte maturation beyond the CD4CD8 stage is The mechanism by which ERK1/2 activation inhibits
reduced in these mice, and the thymocyte subpopulation apoptosis is complicated and varies, depending on the cell
expressing high levels of T cell receptor is similarly decreased, and tissue type involved and the cellular regulatory inuences
624 LU AND XU

Table 1
ERK substrates*
Transcription Kinases and Cytoskeletal Signaling Apoptotic proteins
factors phosphatases proteins proteins and proteinases Other proteins

AML1 (RUNX1) DAPK Annexin XI EGFR Bad Amphiphysin 1


Androgen receptor ERK1/2 Caldesmon ENaCb/g Bim-EL CPSII/CAD
ATF2 FAK1 Calnexin Fe65 Calpain CR16
BCL6 GRK2 CENP-E FRS2 Caspase 9 GRASP55
BMAL1 Inhibitor-2 Connexin-43 Gab1 EDD GRASP65
CBP Lck Cortactin Gab2 IEX1 HABP1
C/EBPb MAPKAP3 Crystallin GAIP MCL-1 Histone H
CRY1/2 MAPKAP5 DOC1R Grb10 TIS2 HnRNP-K
E47 MEK1/2 Dystrophin IRS1 TNFR CD120a KIP
Elk1 MKP1/2 Lamin B2 LAT MBP
ER81 MKP3 MAP1 LIFR PHAS-I (4E-BP1)
ERF MKP7 MAP2 MARCKS CPLA2
Estrogen receptor MLCK MAP4 Naf1a Rb
c-Fos MNK1/2 MISS PDE4 SAP90/PSD95
Fra1 MSK1/2 NF-H PLCb Spinophilin
GATA1/2 PAK1 NF-M PLCg Topoisomerase II
HIF1a PTP2C Paxillin Potassium Tpr
HSF1 Raf1 Stathmin channel Kv 4.2 TTP (Nup47)
ICER B-Raf SWI/SNF KSR1 Tyrosine hydroxylase
c-Jun RSK1-4 Synapsin 1 Rab4 Vif
Microphthalmia S6K Tau SH2-B Vpx
c-Myc Syk Vinexin b ShcA
N-Myc Sos1
Net (Sap2) Spin90
NFATc4 TSC2
NF-IL6
NGFI-B/TR3/Nur77
Pax6
PPARg
P53
Progesterone receptor
RNA Pol. II
PUNX2
Sap1
Smad1
Smad2/3
SP1
SRC1
SREBP1/2
STAT1/3
STAT5a
TAL1/SCL
TFII-I
TFIIIB
TGIF
TIF1A
Tob
UBF

*The more detailed information and more compressive references are available in reference (21).
ROLE OF ERK1/2 MAP KINASES IN CELLS 625

that the cell receives. In Drosophila, MAPK promotes cell cytosols of cells overexpressing B-Raf fails to induce caspase
survival by phosphorylation and inhibition of the pro- activation (47). More direct evidence of inhibited activation of
apoptotic protein head involution defective (Hid) (55), which caspases downstream of cytochrome c has been provided by
activates caspases without involvement of mitochondrial demonstration that caspase-9 is regulated by ERK1/2 (56).
factors (56). In the mammalian cell lines, ERK1/2 signaling Caspase-9 is inhibited by phosphorylation at Thr125 in a
can block apoptosis at levels upstream, downstream, or MEK1/2-dependent manner in cells stimulated with epidermal
unrelated to change of mitochondrial transmembrane poten- growth factor (EGF) or 12-O-tetradecanoylphorbol-13-
tial and cytochrome c release (47, 57, 58). Activation of acetate (TPA). Phosphorylation at Thr125, a conserved
ERK1/2 by constitutive expression of MEK1, or T cell MAP kinase consensus site targeted by ERK2 in vitro, is
activation via T cell receptor stimulation, inhibits caspase 8 sucient to block caspase-9 processing and subsequent
activation/Bid cleavage and the loss of mitochondrial mem- caspase-3 activation (69).
brane potential induced by ionizing radiation or activation of In addition to regulating the activity of survival and
Fas/CD95, TNF, and TRAIL receptors (41, 58, 59). The apoptotic signaling molecules, ERK1/2 can provide an anti-
inactivation of pro-apoptotic Bcl-2 family member BAD is apoptotic eect by up- or down-regulating the protein
mediated through phosphorylation at Ser112 by ERK- expression of such molecules. Exogenous expression of
activated p90 ribosomal S6 kinase (RSK) (60) and at activated Raf-1 enhances the expression of c-Flip, an
Ser136 by phosphoinositol 3-kinase/AKT signaling. BAD endogenous antagonist of caspase-8, in a MEK1/2-dependent
phosphorylation at serine 112 by RSK suppresses BAD- manner (70). Inhibition of ERK1/2 activity causes arrest at
mediated neuronal apoptosis, and inhibition of either ERK1/ the G1 phase of the cell cycle and downregulation of the
2 or AKT increases the extent of apoptosis (61 64). The expression levels of Mcl-1 and Bcl-XL, which are the anti-
ERK-downstream p70 S6 kinase may also be involved in apoptotic homologs of Bcl-2 (71, 72). Increased expression
Ser136 phosphorylation and inhibition of BAD function (65). and activity of DNA repair proteins, such as excision repair
Nerve growth factor (NGF) actively down-regulates the cross-complementing 1 (ERCC1) and X-ray cross-comple-
expression of another pro-apoptotic Bcl-2 family member, menting group 1 (XRCC1), in response to ionizing radiation
Bim, via a MEK-ERK1/2 dependent pathway (66). In have also been found to be dependent upon MEK1/2 activity
addition, NGF promotes rapid MEK-ERK1/2-dependent (73, 74). Furthermore, ERK1/2 facilitates DNA repair by
phosphorylation of Bim at Ser109 and Thr110, thereby regulating the activity of ataxia-telangiectasia mutated
compromising the apoptotic activity of Bim. Thus, MEK- (ATM) and Rad3-related kinase (ATR) (75). By activating
ERK1/2 inhibits Bim activity through two distinct mechan- ERK1/2 via MEK1, hydroxyurea induces MEK-ERK1/2
isms: phosphorylation of Bim and downregulation of Bim dependent S-phase arrest, which can be attenuated by
protein expression (66). inhibition of ERK1/2 activation and thereby sensitize cells
In addition to suppressing the functions of pro-apoptotic to hydroxyurea-induced toxicity. Blocking ERK1/2 activa-
proteins, ERK1/2 can promote cell survival by enhancing tion with MEK inhibitors downregulates ATR function by
the activity of anti-apoptotic molecules. Mcl-1, an anti- reducing the redistribution of ATR from the cytosol to the
apoptotic member of the Bcl-2 family, is phosphorylated at nucleus and the magnitude of formation of ATR nuclear foci
Thr163 by ERK1/2, thus increasing its stability and (75).
enhancing its anti-apoptotic activity (67). IEX-1, an early ERK1/2 controls cell survival or apoptosis by regulating
response and nuclear factor kappa B (NF-kB) target gene, the activity of anti- and pro-apoptotic transcription factors.
interacts with phosphorylated ERK1/2 and is phosphorylated ERK-activated RSK phosphorylates the transcription factor
by ERK1/2 primarily at Thr18 (68). Upon phosphorylation cAMP response element-binding protein (CREB) at serine 133
by ERK1/2, IEX-1 acquires the ability to prevent the (61), which allows recruitment of the co-activators CREB-
release of cytochrome c from mitochondria and to inhibit binding protein (CBP) and p300 and strongly enhances
cell death induced by cytokine deprivation and stimula- CREB-dependent transcription (76). ERK1 can also directly
tion with staurosporine, TNF, or etoposide. In addition to phosphorylate CBP in vitro and increase the transcriptional
acting as an ERK1/2 downstream eector mediating activity of CBP (77). Activated CREB promotes cell survival,
survival, IEX-1 functions as a regulator of ERK1/2 by and inhibition of CREB phosphorylation at serine 133 triggers
enhancing ERK1/2 activation in response to various growth neuronal apoptosis (61). In addition, it has been also shown
factors (68). that inhibition of ERK1/2 upregulates STAT3/5 phosphoryla-
Evidence has shown that ERK1/2 also functions down- tion and promotes cell apoptosis (78).
stream of cytochrome c release. Overexpression of B-Raf, In summary, ERK1/2 can provide anti-apoptotic eects
which does not interfere with the release of cytochrome c from (Fig. 3) by downregulating pro-apoptotic molecules via a
mitochondria, confers MEK1/2 activity-dependent resistance decrease in their activity or a reduction of their protein
to apoptosis induced by growth factor withdrawal or PI expression by transcriptional repression. ERK1/2 can also
3-kinase inhibition; however, the addition of cytochrome c to promote cell survival by upregulating anti-apoptotic molecules
626 LU AND XU

Figure 3. ERK1/2 regulation in their anti-apoptotic function. ERK1/2 activity is positive regulated by MEK1/2 phosphorylation
and negatively regulated by MEKK1-mediated ubiquitination and p38-dependent PP2A activation.

via enhancement of their activity or activation of their


transcription.

Pro-apoptotic Functions of ERK1/2


Although ERK1/2 is a pro-survival factor in the MAP
kinase family and contributes to the regulation of cell
proliferation and dierentiation, under some circumstances,
ERK1/2 can function in a pro-apoptotic manner (Fig. 4). In
the neuronal system, ERK1/2 has been suggested to be
involved in neurodegeneration (79). Inhibition of MEK1/2
confers protection against neuronal cell death induced by the
toxin 6-hydroxydopamine (80). Persistent activation of ERK1/
2 is associated with glutamate-induced oxidative toxicity in
neuronal cells, and inhibition of ERK1/2 activation protects
cells from glutamate toxicity (81). MEK1 inhibition also
results in reduced brain damage and reduced focal infarct
volume in mice after ischemia, which suggests that the MEK1-
ERK1/2 pathway contributes to brain injury during focal
cerebral ischemia (82, 83). In addition, ERK1/2 activation
promotes low potassium-induced neuronal degeneration pre-
dominantly through plasma membrane damage, which occurs Figure 4. Activation of ERK1/2 in response to some DNA
independently of caspase-3 (84). damage stimuli promotes cell apoptosis by enhancing activity
DNA damaging stimuli, including etoposide, adriamycin, of some pro-apoptotic signaling molecules.
platinum compounds, ionizing irradiation, and ultraviolet
irradiation (UV), activate ERK1/2 in primary cells and adriamycin, platinum compounds, or UV. Conversely,
various cell lines (85, 86). Inhibition of ERK1/2 activation enforced activation of ERK1/2 by overexpression of a
attenuates apoptosis or G2-M arrest induced by etoposide, constitutively activated MEK1 Q56P mutant sensitizes cells
ROLE OF ERK1/2 MAP KINASES IN CELLS 627

to DNA damage-induced apoptosis (85). Suppression of the combination of three tiers of kinases, cellular compart-
ERK2 activity has been reported to sensitize ovarian ments in which ERK1/2 is localized, and interaction between
carcinoma cells to cisplatin-induced apoptosis (87, 88), but ERK1/2 and substrates. The specicity of activation or
it has also been reported to induce drug resistance of cervical inhibition of downstream eectors determines the consequence
carcinoma cells to cisplatin (89, 90) and various other tumor of ERK1/2 activation on cell survival, which is anti-apoptotic
cells to multiple anticancer drugs (85, 91, 92). Inhibition of but in some cases pro-apoptotic. Further elucidation of how
MEK-ERK1/2 activity also decreases isothiocyanates- and ERK1/2 executes its function through downstream factors will
20 -hydroxycinnamaldehyde-induced apoptosis and cell help us understand completely ERK1/2 function in cell
growth (93, 94). survival and apoptosis.
How ERK1/2 activation promotes apoptosis is not well
understood. In radiation-induced apoptosis, PKC d enhances
the cytotoxic eect by allowing cells to escape from radiation- ACKNOWLEDGMENTS
induced arrest in the G2-M phases (69). However, this eect is We thank Oliver Bogler, Pamela Woodring, David Giegel,
reduced when radiation-induced ERK1/2 activity is inhibited John de Groot and Theresa Willis for their critical reading of
by a MEK inhibitor (95). Treatment of human leukemia U937 the manuscript and Dexing Fang and Yanhua Zheng for
cells with EGF activates ERK1/2, which in turn phosphor- preparation of the gures and table.
ylates Ser147 of tis21 and induces the interaction between tis21 This work was supported by National Cancer Institute
and Pin-1, mitochondrial depolarization, G2-M arrest, and grant 1R01CA109035-01A1 (Z.L.), the Pediatric Brain Tumor
cell apoptosis (96). Inhibition of ERK1/2 activity by MEK Foundation (Z.L.), the Charlotte Geyer Foundation (Z.L.),
inhibitors or a dominant-negative mutant of MEK1 reduces and an institutional research grant from The University of
cisplatin-induced increases in Bax expression, mitochondrial Texas M. D. Anderson Cancer Center (Z.L.).
membrane depolarization, mitochondrial cytochrome c re-
lease, caspase-3 activation, and cell apoptosis (97 99). REFERENCES
Doxorubicin activates ERK2, which in turn associates with 1. Rubin, G. M., Yandell, M. D., Wortman, J. R., Gabor Miklos, G.
and phosphorylates p53 at Thr55, thereby increasing p53 L., Nelson, C. R., Hariharan, I. K., Fortini, M. E., Li, P. W.,
Apweiler, R., Fleischmann, W., Cherry, J. M., Heniko, S., Skupski,
transcriptional activity (100). Overexpression of p53T55A
M. P., Misra, S., Ashburner, M., Birney, E., Boguski, M. S., Brody,
suppresses the activity of endogenous p53 in MCF7 breast T., Brokstein, P., Celniker, S. E., Chervitz, S. A., Coates, D.,
cancer cells, enhances expression level of Bcl-2, and signi- Cravchik, A., Gabrielian, A., Galle, R. F., Gelbart, W. M., George,
cantly increases drug resistance toward doxorubicin (92). R. A., Goldstein, L. S., Gong, F., Guan, P., Harris, N. L., Hay, B.
Cisplatin-induced phosphorylation of p53 at serine 15, A., Hoskins, R. A., Li, J., Li, Z., Hynes, R. O., Jones, S. J., Kuehl, P.
M., Lemaitre, B., Littleton, J. T., Morrison, D. K., Mungall, C.,
increase of p53 protein half-life, and accumulation of p53
OFarrell, P. H., Pickeral, O. K., Shue, C., Vosshall, L. B., Zhang, J.,
downstream gene expression such as p21WAF-1 and MDM2 in Zhao, Q., Zheng, X. H., and Lewis, S. (2000) Comparative genomics
A2780 ovarian carcinoma cells are blocked by inhibition of of the eukaryotes. Science 287, 2204 2215.
ERK1/2 (88). Nevertheless, ERK-independent regulation of 2. Lander, E. S., Linton, L. M., Birren, B., Nusbaum, C., Zody, M. C.,
p53 was also reported. ERK activation increases cisplatin- Baldwin, J., Devon, K., Dewar, K., Doyle, M., FitzHugh, W., Funke, R.,
Gage, D., Harris, K., Heaford, A., Howland, J., Kann, L., Lehoczky, J.,
induced cell death in osteosarcoma and neuroblastoma cell
LeVine, R., McEwan, P., McKernan, K., Meldrim, J., Mesirov, J. P.,
lines (91) and mediates cell cycle arrest and apoptosis in Miranda, C., Morris, W., Naylor, J., Raymond, C., Rosetti, M., Santos,
response to etoposide, adriamycin, and ionizing irradiation R., Sheridan, A., Sougnez, C., Stange-Thomann, N., Stojanovic, N.,
(85) in a p53-independent manner. Thus, upregulation of p53 Subramanian, A., Wyman, D., Rogers, J., Sulston, J., Ainscough, R.,
activity by ERK activation during apoptosis appears to be Beck, S., Bentley, D., Burton, J., Clee, C., Carter, N., Coulson, A.,
Deadman, R., Deloukas, P., Dunham, A., Dunham, I., Durbin, R.,
dependent on types of cell lines and stimuli, and the
French, L., Grafham, D., Gregory, S., Hubbard, T., Humphray, S.,
mechanism by which ERK1/2 activation promotes apoptosis Hunt, A., Jones, M., Lloyd, C., McMurray, A., Matthews, L., Mercer,
warrants further investigation. S., Milne, S., Mullikin, J. C., Mungall, A., Plumb, R., Ross, M.,
Shownkeen, R., Sims, S., Waterston, R. H., Wilson, R. K., Hillier, L. W.,
McPherson, J. D., Marra, M. A., Mardis, E. R., Fulton, L. A.,
CONCLUSIONS Chinwalla, A. T., Pepin, K. H., Gish, W. R., Chissoe, S. L., Wendl, M. C.,
Delehaunty, K. D., Miner, T. L., Delehaunty, A., Kramer, J. B., Cook, L.
The ERK1/2 cascade is a central signaling pathway L., Fulton, R. S., Johnson, D. L., Minx, P. J., Clifton, S. W., Hawkins, T.,
responsive to a large number of extracellular stimuli. ERK1/ Branscomb, E., Predki, P., Richardson, P., Wenning, S., Slezak, T.,
2 can be activated transiently or persistently by dierent Doggett, N., Cheng, J. F., Olsen, A., Lucas, S., Elkin, C., Uberbacher, E.,
MAP3Ks in conjunction with involvement of various scaold- Frazier, M., Gibbs, R. A., Muzny, D. M., Scherer, S. E., Bouck, J. B.,
Sodergren, E. J., Worley, K. C., Rives, C. M., Gorrell, J. H., Metzker, M.
ing proteins and phosphatases. Activated ERK1/2 executes its
L., Naylor, S. L., Kucherlapati, R. S., Nelson, D. L., Weinstock, G. M.,
function through a large number of downstream molecules. Sakaki, Y., Fujiyama, A., Hattori, M., Yada, T., Toyoda, A., Itoh, T.,
The targeted selection of ERK1/2 downstream substrates Kawagoe, C., Watanabe, H., Totoki, Y., Taylor, T., Weissenbach, J.,
depends upon the cell type, nature of the stimuli that specify Heilig, R., Saurin, W., Artiguenave, F., Brottier, P., Bruls, T.,
628 LU AND XU

Pelletier, E., Robert, C., Wincker, P., Smith, D. R., Doucette-Stamm, L., 17. Holland, P. M., and Cooper, J. A. (1999) Protein modication:
Rubeneld, M., Weinstock, K., Lee, H. M., Dubois, J., Rosenthal, A., docking sites for kinases. Curr. Biol. 9, R329 331.
Platzer, M., Nyakatura, G., Taudien, S., Rump, A., Yang, H., Yu, J., 18. Fantz, D. A., Jacobs, D., Glossip, D., and Kornfeld, K. (2001)
Wang, J., Huang, G., Gu, J., Hood, L., Rowen, L., Madan, A., Qin, S., Docking sites on substrate proteins direct extracellular signal-
Davis, R. W., Federspiel, N. A., Abola, A. P., Proctor, M. J., Myers, R. regulated kinase to phosphorylate specic residues. J. Biol. Chem.
M., Schmutz, J., Dickson, M., Grimwood, J., Cox, D. R., Olson, M. V., 276, 27256 27265.
Kaul, R., Shimizu, N., Kawasaki, K., Minoshima, S., Evans, G. A., 19. Jacobs, D., Glossip, D., Xing, H., Muslin, A. J., and Kornfeld, K.
Athanasiou, M., Schultz, R., Roe, B. A., Chen, F., Pan, H., Ramser, J., (1999) Multiple docking sites on substrate proteins form a modular
Lehrach, H., Reinhardt, R., McCombie, W. R., de la Bastide, M., system that mediates recognition by ERK MAP kinase. Genes Dev.
Dedhia, N., Blocker, H., Hornischer, K., Nordsiek, G., Agarwala, R., 13, 163 175.
Aravind, L., Bailey, J. A., Bateman, A., Batzoglou, S., Birney, E., 20. MacKenzie, S. J., Baillie, G. S., McPhee, I., Bolger, G. B., and
Bork, P., Brown, D. G., Burge, C. B., Cerutti, L., Chen, H. C., Houslay, M. D. (2000) ERK2 mitogen-activated protein kinase
Church, D., Clamp, M., Copley, R. R., Doerks, T., Eddy, S. R., binding, phosphorylation, and regulation of the PDE4D cAMP-
Eichler, E. E., Furey, T. S., Galagan, J., Gilbert, J.G., Harmon, C., specic phosphodiesterases. The involvement of COOH-terminal
Hayashizaki, Y., Haussler, D., Hermjakob, H., Hokamp, K., Jang, W., docking sites and NH2-terminal UCR regions. J. Biol. Chem. 275,
Johnson, L. S., Jones, T. A., Kasif, S., Kaspryzk, A., Kennedy, S., 16609 16617.
Kent, W.J., Kitts, P., Koonin, E.V., Korf, I., Kulp, D., Lancet, D., 21. Lee, T., Hoofnagle, A. N., Kabuyama, Y., Stroud, J., Min, X.,
Lowe, T. M., McLysaght, A., Mikkelsen, T., Moran, J.V., Mulder, N., Goldsmith, E. J., Chen, L., Resing, K. A., and Ahn, N. G. (2004)
Pollara, V. J., Ponting, C. P., Schuler, G., Schultz, J., Slater, G., Docking motif interactions in MAP kinases revealed by hydrogen
Smit, A. F., Stupka, E., Szustakowski, J., Thierry-Mieg, D., Thierry- exchange mass spectrometry. Mol. Cell 14, 43 55.
Mieg, J., Wagner, L., Wallis, J., Wheeler, R., Williams, A., Wolf, Y. I., 22. Yoon, S., and Seger, R. (2006) The extracellular signal-regulated
Wolfe, K. H., Yang, S. P., Yeh, R. F., Collins, F., Guyer, M. S., kinase: multiple substrates regulate diverse cellular functions. Growth
Peterson, J., Felsenfeld, A., Wetterstrand, K. A., Patrinos, A., Factors 24, 21 44.
Morgan, M. J., de Jong, P., Catanese, J. J., Osoegawa, K., 23. Boulton, T. G., Nye, S. H., Robbins, D. J., Ip, N. Y., Radziejewska, E.,
Shizuya, H., Choi, S., and Chen, Y. J. (2001) Initial sequencing and Morgenbesser, S. D., DePinho, R. A., Panayotatos, N., Cobb, M. H.,
analysis of the human genome. Nature 409, 860 921. and Yancopoulos, G. D. (1991) ERKs: a family of protein-serine/
3. Manning, G., Whyte, D. B., Martinez, R., Hunter, T., and threonine kinases that are activated and tyrosine phosphorylated in
Sudarsanam, S. (2002) The protein kinase complement of the human response to insulin and NGF. Cell 65, 663 675.
genome. Science 298, 1912 1934. 24. Chadee, D. N., and Kyriakis, J. M. (2004) MLK3 is required for
4. Widmann, C., Gibson, S., Jarpe, M. B., and Johnson, G. L. (1999) mitogen activation of B-Raf, ERK and cell proliferation. Nat. Cell
Mitogen-activated protein kinase: conservation of a three-kinase Biol. 6, 770 776.
module from yeast to human. Physiol. Rev. 79, 143 180. 25. Gotoh, Y., and Nishida, E. (1995) Activation mechanism and function
5. Chen, Z., Gibson, T. B., Robinson, F., Silvestro, L., Pearson, G., of the MAP kinase cascade. Mol. Reprod. Dev. 42, 486 492.
Xu, B., Wright, A., Vanderbilt, C., and Cobb, M. H. (2001) MAP 26. Salmeron, A., Ahmad, T. B., Carlile, G. W., Pappin, D., Narsimhan, R.
kinases. Chem. Rev. 101, 2449 2476. P., and Ley, S. C. (1996) Activation of MEK-1 and SEK-1 by Tpl-2
6. Platanias, L. C. (2003) Map kinase signaling pathways and proto-oncoprotein, a novel MAP kinase kinase kinase. Embo J. 15,
hematologic malignancies. Blood 101, 4667 4679. 817 826.
7. Bogoyevitch, M. A., and Court, N. W. (2004) Counting on mitogen- 27. Gotoh, I., Adachi, M., and Nishida, E. (2001) Identication and
activated protein kinases ERKs 3, 4, 5, 6, 7 and 8. Cell Signal 16, characterization of a novel MAP kinase kinase kinase, MLTK.
1345 1354. J. Biol. Chem. 276, 4276 4286.
8. Chang, L., and Karin, M. (2001) Mammalian MAP kinase signalling 28. MacGillivray, M. K., Cruz, T. F., and McCulloch, C. A. (2000)
cascades. Nature 410, 37 40. The recruitment of the interleukin-1 (IL-1) receptor-associated
9. Leppa, S., and Bohmann, D. (1999) Diverse functions of JNK signaling kinase (IRAK) into focal adhesion complexes is required for
and c-Jun in stress response and apoptosis. Oncogene 18, 6158 6162. IL-1beta -induced ERK activation. J. Biol. Chem. 275, 23509
10. Raman, M., and Cobb, M. H. (2003) MAP kinase modules: many 23515.
roads home. Curr. Biol. 13, R886 888. 29. Blank, J. L., Gerwins, P., Elliott, E. M., Sather, S., and Johnson, G. L.
11. Ge, B., Gram, H., Di Padova, F., Huang, B., New, L., Ulevitch, R. (1996) Molecular cloning of mitogen-activated protein/ERK kinase
J., Luo, Y., and Han, J. (2002) MAPKK-independent activation of kinases (MEKK) 2 and 3. Regulation of sequential phosphorylation
p38alpha mediated by TAB1-dependent autophosphorylation of pathways involving mitogen-activated protein kinase and c-Jun
p38alpha. Science 295, 1291 1294. kinase. J. Biol. Chem. 271, 5361 5368.
12. Chen, W., White, M. A., and Cobb, M. H. (2002) Stimulus-specic 30. Karandikar, M., Xu, S., and Cobb, M. H. (2000) MEKK1 binds raf-
requirements for MAP3 kinases in activating the JNK pathway. 1 and the ERK2 cascade components. J. Biol. Chem. 275, 40120
J. Biol. Chem. 277, 49105 49110. 40127.
13. Kolch, W. (2005) Coordinating ERK/MAPK signalling through 31. Waetzig, V., and Herdegen, T. (2005) MEKK1 controls neurite
scaolds and inhibitors. Nat. Rev. Mol. Cell Biol. 6, 827 837. regrowth after experimental injury by balancing ERK1/2 and JNK2
14. Tanoue, T., Adachi, M., Moriguchi, T., and Nishida, E. (2000) A signaling. Mol. Cell Neurosci. 30, 67 78.
conserved docking motif in MAP kinases common to substrates, 32. Lange-Carter, C. A., Pleiman, C. M., Gardner, A. M., Blumer, K. J.,
activators and regulators. Nat. Cell Biol. 2, 110 116. and Johnson, G. L. (1993) A divergence in the MAP kinase
15. Tanoue, T., Maeda, R., Adachi, M., and Nishida, E. (2001) Identica- regulatory network dened by MEK kinase and Raf. Science 260,
tion of a docking groove on ERK and p38 MAP kinases that regulates 315 319.
the specicity of docking interactions. Embo J. 20, 466 479. 33. Lu, Z., Xu, S., Joazeiro, C., Cobb, M. H., and Hunter, T. (2002)
16. Sharrocks, A. D., Yang, S. H., and Galanis, A. (2000) Docking The PHD domain of MEKK1 acts as an E3 ubiquitin ligase and
domains and substrate-specicity determination for MAP kinases. mediates ubiquitination and degradation of ERK1/2. Mol. Cell 9,
Trends Biochem. Sci. 25, 448 453. 945 956.
ROLE OF ERK1/2 MAP KINASES IN CELLS 629

34. Gonzalez, F. A., Raden, D. L., and Davis, R. J. (1991) Identication 47. Erhardt, P., Schremser, E. J., and Cooper, G. M. (1999) B-Raf
of substrate recognition determinants for human ERK1 and ERK2 inhibits programmed cell death downstream of cytochrome c release
protein kinases. J. Biol. Chem. 266, 22159 22163. from mitochondria by activating the MEK/Erk pathway. Mol. Cell.
35. Songyang, Z., Lu, K. P., Kwon, Y. T., Tsai, L. H., Filhol, O., Cochet, C., Biol. 19, 5308 5315.
Brickey, D. A., Soderling, T. R., Bartleson, C., Graves, D. J., DeMaggio, 48. Kim, S. J., Ju, J. W., Oh, C. D., Yoon, Y. M., Song, W. K., Kim, J.
A. J., Hoekstra, M. F., Blenis, J., Hunter, T., and Cantley, L. C. (1996) H., Yoo, Y. J., Bang, O. S., Kang, S. S., and Chun, J. S. (2002) ERK-
A structural basis for substrate specicities of protein Ser/Thr kinases: 1/2 and p38 kinase oppositely regulate nitric oxide-induced apoptosis
primary sequence preference of casein kinases I and II, NIMA, of chondrocytes in association with p53, caspase-3, and dierentia-
phosphorylase kinase, calmodulin-dependent kinase II, CDK5, and tion status. J. Biol. Chem. 277, 1332 1339.
Erk1. Mol. Cell Biol. 16, 6486 6493. 49. Wang, X., Martindale, J. L., Liu, Y., and Holbrook, N. J. (1998) The
36. Pages, G., Guerin, S., Grall, D., Bonino, F., Smith, A., Anjuere, F., cellular response to oxidative stress: inuences of mitogen-activated
Auberger, P., and Pouyssegur, J. (1999) Defective thymocyte protein kinase signalling pathways on cell survival. Biochem. J. 333
maturation in p44 MAP kinase (Erk 1) knockout mice. Science (Pt 2), 291 300.
286, 1374 1377. 50. Le Gall, M., Chambard, J. C., Breittmayer, J. P., Grall, D.,
37. Mazzucchelli, C., Vantaggiato, C., Ciamei, A., Fasano, S., Pakhotin, Pouyssegur, J., and Van Obberghen-Schilling, E. (2000) The p42/p44
P., Krezel, W., Welzl, H., Wolfer, D. P., Pages, G., Valverde, O., MAP kinase pathway prevents apoptosis induced by anchorage and
Marowsky, A., Porrazzo, A., Orban, P. C., Maldonado, R., serum removal. Mol. Biol. Cell. 11, 1103 1112.
Ehrengruber, M. U., Cestari, V., Lipp, H. P., Chapman, P. F., 51. Anderson, C. N., and Tolkovsky, A. M. (1999) A role for MAPK/
Pouyssegur, J., and Brambilla, R. (2002) Knockout of ERK1 MAP ERK in sympathetic neuron survival: protection against a p53-
kinase enhances synaptic plasticity in the striatum and facilitates dependent, JNK-independent induction of apoptosis by cytosine
striatal-mediated learning and memory. Neuron 34, 807 820. arabinoside. J. Neurosci. 19, 664 673.
38. Yao, Y., Li, W., Wu, J., Germann, U. A., Su, M. S., Kuida, K., and 52. Lee, J., Hong, F., Kwon, S., Kim, S. S., Kim, D. O., Kang, H. S.,
Boucher, D. M. (2003) Extracellular signal-regulated kinase 2 is Lee, S. J., Ha, J., and Kim, S. S. (2002) Activation of p38 MAPK
necessary for mesoderm dierentiation. Proc. Natl. Acad. Sci. USA induces cell cycle arrest via inhibition of Raf/ERK pathway during
100, 12759 12764. muscle dierentiation. Biochem. Biophys. Res. Commun. 298, 765
39. Hatano, N., Mori, Y., Oh-hora, M., Kosugi, A., Fujikawa, T., 771.
Nakai, N., Niwa, H., Miyazaki, J., Hamaoka, T., and Ogata, M. 53. Li, S. P., Junttila, M. R., Han, J., Kahari, V. M., and Westermarck,
(2003) Essential role for ERK2 mitogen-activated protein kinase in J. (2003) p38 Mitogen-activated protein kinase pathway suppresses
placental development. Genes Cells 8, 847 856. cell survival by inducing dephosphorylation of mitogen-activated
40. Saba-El-Leil, M. K., Vella, F. D., Vernay, B., Voisin, L., Chen, L., protein/extracellular signal-regulated kinase kinase1,2. Cancer Res.
Labrecque, N., Ang, S. L., and Meloche, S. (2003) An essential 63, 3473 3477.
function of the mitogen-activated protein kinase Erk2 in mouse 54. Liu, Q., and Hofmann, P. A. (2004) Protein phosphatase 2A-
trophoblast development. EMBO Rep. 4, 964 968. mediated cross-talk between p38 MAPK and ERK in apoptosis of
41. Tran, S. E., Holmstrom, T. H., Ahonen, M., Kahari, V. M., and cardiac myocytes. Am. J. Physiol. Heart Circ. Physiol. 286, H2204
Eriksson, J. E. (2001) MAPK/ERK overrides the apoptotic signaling 2212.
from Fas, TNF, and TRAIL receptors. J. Biol. Chem. 276, 16484 55. Bergmann, A., Agapite, J., McCall, K., and Steller, H. (1998) The
16490. Drosophila gene hid is a direct molecular target of Ras-dependent
42. Park, J. S., Carter, S., Reardon, D. B., Schmidt-Ullrich, R., Dent, P., survival signaling. Cell 95, 331 341.
and Fisher, P. B. (1999) Roles for basal and stimulated p21(Cip-1/ 56. Means, J. C., Muro, I., and Clem, R. J. (2006) Lack of involvement
WAF1/MDA6) expression and mitogen-activated protein kinase of mitochondrial factors in caspase activation in a Drosophila cell-
signaling in radiation-induced cell cycle checkpoint control in free system. Cell Death Dier. 13, 1222 1234.
carcinoma cells. Mol. Biol. Cell. 10, 4231 4246. 57. Qiao, L., Studer, E., Leach, K., McKinstry, R., Gupta, S., Decker,
43. Kitagawa, D., Tanemura, S., Ohata, S., Shimizu, N., Seo, J., R., Kukreja, R., Valerie, K., Nagarkatti, P., El Deiry, W.,
Nishitai, G., Watanabe, T., Nakagawa, K., Kishimoto, H., Wada, Molkentin, J., Schmidt-Ullrich, R., Fisher, P. B., Grant, S.,
T., Tezuka, T., Yamamoto, T., Nishina, H., and Katada, T. (2002) Hylemon, P. B., and Dent, P. (2001) Deoxycholic acid (DCA)
Activation of extracellular signal-regulated kinase by ultraviolet is causes ligand-independent activation of epidermal growth factor
mediated through Src-dependent epidermal growth factor receptor receptor (EGFR) and FAS receptor in primary hepatocytes:
phosphorylation. Its implication in an anti-apoptotic function. inhibition of EGFR/mitogen-activated protein kinase-signaling
J. Biol. Chem. 277, 366 371. module enhances DCA-induced apoptosis. Mol. Biol. Cell 12,
44. Foncea, R., Galvez, A., Perez, V., Morales, M. P., Calixto, A., 2629 2645.
Melendez, J., Gonzalez-Jara, F., Diaz-Araya, G., Sapag-Hagar, M., 58. Shonai, T., Adachi, M., Sakata, K., Takekawa, M., Endo, T., Imai,
Sugden, P. H., LeRoith, D., and Lavandero, S. (2000) Extracellular K., and Hareyama, M. (2002) MEK/ERK pathway protects
regulated kinase, but not protein kinase C, is an antiapoptotic signal ionizing radiation-induced loss of mitochondrial membrane potential
of insulin-like growth factor-1 on cultured cardiac myocytes. and cell death in lymphocytic leukemia cells. Cell Death Dier. 9,
Biochem. Biophys. Res. Commun. 273, 736 744. 963 971.
45. Nagata, Y., and Todokoro, K. (1999) Requirement of activation of 59. Holmstrom, T. H., Schmitz, I., Soderstrom, T. S., Poukkula, M.,
JNK and p38 for environmental stress-induced erythroid dierentia- Johnson, V. L., Chow, S. C., Krammer, P. H., and Eriksson, J. E.
tion and apoptosis and of inhibition of ERK for apoptosis. Blood 94, (2000) MAPK/ERK signaling in activated T cells inhibits CD95/Fas-
853 863. mediated apoptosis downstream of DISC assembly. Embo J. 19,
46. Buckley, S., Driscoll, B., Barsky, L., Weinberg, K., Anderson, K., 5418 5428.
and Warburton, D. (1999) ERK activation protects against DNA 60. Sturgill, T. W., Ray, L. B., Erikson, E., and Maller, J. L. (1988)
damage and apoptosis in hyperoxic rat AEC2. Am. J. Physiol. 277, Insulin-stimulated MAP-2 kinase phosphorylates and activates
L159 166. ribosomal protein S6 kinase II. Nature 334, 715 718.
630 LU AND XU

61. Bonni, A., Brunet, A., West, A. E., Datta, S. R., Takasu, M. A., and 77. Janknecht, R., and Nordheim, A. (1996) MAP kinase-dependent
Greenberg, M. E. (1999) Cell survival promoted by the Ras-MAPK transcriptional coactivation by Elk-1 and its cofactor CBP. Biochem.
signaling pathway by transcription-dependent and -independent Biophys. Res. Commun. 228, 831 837.
mechanisms. Science 286, 1358 1362. 78. Krasilnikov, M., Ivanov, V. N., Dong, J., and Ronai, Z. (2003) ERK
62. Scheid, M. P., Schubert, K. M., and Duronio, V. (1999) Regulation and PI3K negatively regulate STAT-transcriptional activities in
of bad phosphorylation and association with Bcl-x(L) by the MAPK/ human melanoma cells: implications towards sensitization to
Erk kinase. J. Biol. Chem. 274, 31108 31113. apoptosis. Oncogene 22, 4092 4101.
63. Hayakawa, J., Ohmichi, M., Kurachi, H., Kanda, Y., Hisamoto, K., 79. Cheung, E. C., and Slack, R. S. (2004) Emerging role for ERK as a
Nishio, Y., Adachi, K., Tasaka, K., Kanzaki, T., and Murata, Y. key regulator of neuronal apoptosis. Sci. STKE 2004, PE45.
(2000) Inhibition of BAD phosphorylation either at serine 112 via 80. Kulich, S. M., and Chu, C. T. (2001) Sustained extracellular signal-
extracellular signal-regulated protein kinase cascade or at serine 136 regulated kinase activation by 6-hydroxydopamine: implications for
via Akt cascade sensitizes human ovarian cancer cells to cisplatin. Parkinsons disease. J. Neurochem. 77, 1058 1066.
Cancer Res. 60, 5988 5994. 81. Stanciu, M., Wang, Y., Kentor, R., Burke, N., Watkins, S., Kress, G.,
64. Mabuchi, S., Ohmichi, M., Kimura, A., Hisamoto, K., Hayakawa, Reynolds, I., Klann, E., Angiolieri, M. R., Johnson, J. W., and
J., Nishio, Y., Adachi, K., Takahashi, K., Arimoto-Ishida, E., DeFranco, D. B. (2000) Persistent activation of ERK contributes
Nakatsuji, Y., Tasaka, K., and Murata, Y. (2002) Inhibition of to glutamate-induced oxidative toxicity in a neuronal cell line
phosphorylation of BAD and Raf-1 by Akt sensitizes human ovarian and primary cortical neuron cultures. J. Biol. Chem. 275, 12200
cancer cells to paclitaxel. J. Biol. Chem. 277, 33490 33500. 12206.
65. Harada, H., Andersen, J. S., Mann, M., Terada, N., and Korsmeyer, 82. Alessandrini, A., Namura, S., Moskowitz, M. A., and Bonventre, J. V.
S. J. (2001) p70S6 kinase signals cell survival as well as growth, (1999) MEK1 protein kinase inhibition protects against damage
inactivating the pro-apoptotic molecule BAD. Proc. Natl. Acad. Sci. resulting from focal cerebral ischemia. Proc. Natl. Acad. Sci. USA 96,
USA 98, 9666 9670. 12866 12869.
66. Biswas, S. C., and Greene, L. A. (2002) Nerve growth factor (NGF) 83. Namura, S., Iihara, K., Takami, S., Nagata, I., Kikuchi, H.,
down-regulates the Bcl-2 homology 3 (BH3) domain-only protein Matsushita, K., Moskowitz, M. A., Bonventre, J. V., and
Bim and suppresses its proapoptotic activity by phosphorylation. Alessandrini, A. (2001) Intravenous administration of MEK
J. Biol. Chem. 277, 49511 49516. inhibitor U0126 aords brain protection against forebrain ischemia
67. Domina, A. M., Vrana, J. A., Gregory, M. A., Hann, S. R., and and focal cerebral ischemia. Proc. Natl. Acad. Sci. USA 98, 11569
Craig, R. W. (2004) MCL1 is phosphorylated in the PEST region and 11574.
stabilized upon ERK activation in viable cells, and at additional sites 84. Subramaniam, S., Zirrgiebel, U., von Bohlen Und Halbach, O.,
with cytotoxic okadaic acid or taxol. Oncogene 23, 5301 5315. Strelau, J., Laliberte, C., Kaplan, D. R., and Unsicker, K. (2004)
68. Garcia, J., Ye, Y., Arranz, V., Letourneux, C., Pezeron, G., and ERK activation promotes neuronal degeneration predominantly
Porteu, F. (2002) IEX-1: a new ERK substrate involved in both ERK through plasma membrane damage and independently of caspase-3.
survival activity and ERK activation. Embo J. 21, 5151 5163. J. Cell Biol. 165, 357 369.
69. Allan, L. A., Morrice, N., Brady, S., Magee, G., Pathak, S., and 85. Tang, D., Wu, D., Hirao, A., Lahti, J. M., Liu, L., Mazza, B., Kidd,
Clarke, P. R. (2003) Inhibition of caspase-9 through phosphorylation V. J., Mak, T. W., and Ingram, A. J. (2002) ERK activation mediates
at Thr 125 by ERK MAPK. Nat. Cell Biol. 5, 647 654. cell cycle arrest and apoptosis after DNA damage independently of
70. Aoudjit, F., and Vuori, K. (2001) Matrix attachment regulates Fas- p53. J. Biol. Chem. 277, 12710 12717.
induced apoptosis in endothelial cells: a role for c-ip and 86. Billecke, C., Finniss, S., Tahash, L., Miller, C., Mikkelsen, T.,
implications for anoikis. J. Cell Biol. 152, 633 643. Farrell, N. P., and Bogler, O. (2006) Polynuclear platinum anticancer
71. Boucher, M. J., Morisset, J., Vachon, P. H., Reed, J. C., Laine, J., drugs are more potent than cisplatin and induce cell cycle arrest in
and Rivard, N. (2000) MEK/ERK signaling pathway regulates the glioma. Neuro-oncol. 8, 215 226.
expression of Bcl-2, Bcl-X(L), and Mcl-1 and promotes survival of 87. Hayakawa, J., Ohmichi, M., Kurachi, H., Ikegami, H., Kimura, A.,
human pancreatic cancer cells. J. Cell Biochem. 79, 355 369. Matsuoka, T., Jikihara, H., Mercola, D., and Murata, Y. (1999)
72. Jost, M., Huggett, T. M., Kari, C., Boise, L. H., and Rodeck, U. Inhibition of extracellular signal-regulated protein kinase or c-Jun
(2001) Epidermal growth factor receptor-dependent control of N-terminal protein kinase cascade, dierentially activated by
keratinocyte survival and Bcl-xL expression through a MEK- cisplatin, sensitizes human ovarian cancer cell line. J. Biol. Chem.
dependent pathway. J. Biol. Chem. 276, 6320 6326. 274, 31648 31654.
73. Yacoub, A., Park, J. S., Qiao, L., Dent, P., and Hagan, M. P. (2001) 88. Persons, D. L., Yazlovitskaya, E. M., and Pelling, J. C. (2000) Eect
MAPK dependence of DNA damage repair: ionizing radiation and of extracellular signal-regulated kinase on p53 accumulation in
the induction of expression of the DNA repair genes XRCC1 and response to cisplatin. J. Biol. Chem. 275, 35778 35785.
ERCC1 in DU145 human prostate carcinoma cells in a MEK1/2 89. Wang, X., Martindale, J. L., and Holbrook, N. J. (2000) Require-
dependent fashion. Int. J. Radiat. Biol. 77, 1067 1078. ment for ERK activation in cisplatin-induced apoptosis. J. Biol.
74. Yacoub, A., McKinstry, R., Hinman, D., Chung, T., Dent, P., and Chem. 275, 39435 39443.
Hagan, M. P. (2003) Epidermal growth factor and ionizing radiation 90. Yeh, P. Y., Chuang, S. E., Yeh, K. H., Song, Y. C., Ea, C. K., and
up-regulate the DNA repair genes XRCC1 and ERCC1 in DU145 Cheng, A. L. (2002) Increase of the resistance of human cervical
and LNCaP prostate carcinoma through MAPK signaling. Radiat. carcinoma cells to cisplatin by inhibition of the MEK to ERK
Res. 159, 439 452. signaling pathway partly via enhancement of anticancer drug-
75. Wu, D., Chen, B., Parihar, K., He, L., Fan, C., Zhang, J., Liu, L., induced NF kappa B activation. Biochem. Pharmacol. 63, 1423
Gillis, A., Bruce, A., Kapoor, A., and Tang, D. (2006) ERK activity 1430.
facilitates activation of the S-phase DNA damage checkpoint by 91. Woessmann, W., Chen, X., and Borkhardt, A. (2002) Ras-mediated
modulating ATR function. Oncogene 25, 1153 1164. activation of ERK by cisplatin induces cell death independently of
76. Johannessen, M., Delghandi, M. P., and Moens, U. (2004) What p53 in osteosarcoma and neuroblastoma cell lines. Cancer
turns CREB on? Cell Signal 16, 1211 1227. Chemother. Pharmacol. 50, 397 404.
ROLE OF ERK1/2 MAP KINASES IN CELLS 631

92. Yeh, P. Y., Chuang, S. E., Yeh, K. H., Song, Y. C., Chang, L. L., 96. Hong, J. W., Ryu, M. S., and Lim, I. K. (2005) Phosphorylation of
and Cheng, A. L. (2004) Phosphorylation of p53 on Thr55 by ERK2 serine 147 of tis21/BTG2/pc3 by p-Erk1/2 induces Pin-1 binding in
is necessary for doxorubicin-induced p53 activation and cell death. cytoplasm and cell death. J. Biol. Chem. 280, 21256 21263.
Oncogene 23, 3580 3588. 97. Schweyer, S., Soruri, A., Meschter, O., Heintze, A., Zschunke, F.,
93. Xu, C., Shen, G., Yuan, X., Kim, J. H., Gopalkrishnan, A., Keum, Miosge, N., Thelen, P., Schlott, T., Radzun, H. J., and Fayyazi, A.
Y. S., Nair, S., and Kong, A. N. (2006) ERK and JNK signaling (2004) Cisplatin-induced apoptosis in human malignant testicular
pathways are involved in the regulation of activator protein 1 and germ cell lines depends on MEK/ERK activation. Br. J. Cancer 91,
cell death elicited by three isothiocyanates in human prostate cancer 589 598.
PC-3 cells. Carcinogenesis 27, 437 445. 98. Kim, Y. K., Kim, H. J., Kwon, C. H., Kim, J. H., Woo, J. S., Jung, J. S.,
94. Lee, S. H., Lee, C. W., Lee, J. W., Choi, M. S., Son, D. J., Chung, Y. and Kim, J. M. (2005) Role of ERK activation in cisplatin-induced
B., Lee, C. K., Oh, K. W., Moon, D. C., Kwon, B. M., and Hong, apoptosis in OK renal epithelial cells. J. Appl. Toxicol. 25, 374 382.
J. T. (2005) Induction of apoptotic cell death by 20 -hydroxycinnam- 99. Jo, S. K., Cho, W. Y., Sung, S. A., Kim, H. K., and Won, N. H.
aldehyde is involved with ERK-dependent inactivation of NF- (2005) MEK inhibitor, U0126, attenuates cisplatin-induced renal
kappaB in TNF-alpha-treated SW620 colon cancer cells. Biochem. injury by decreasing inammation and apoptosis. Kidney Int. 67,
Pharmacol. 70, 1147 1157. 458 466.
95. Lee, Y. J., Soh, J. W., Dean, N. M., Cho, C. K., Kim, T. H., Lee, S. 100. Yeh, P. Y., Chuang, S. E., Yeh, K. H., Song, Y. C., and Cheng, A. L.
J., and Lee, Y. S. (2002) Protein kinase Cdelta overexpression (2001) Nuclear extracellular signal-regulated kinase 2 phosphorylates
enhances radiation sensitivity via extracellular regulated protein p53 at Thr55 in response to doxorubicin. Biochem. Biophys. Res.
kinase 1/2 activation, abolishing the radiation-induced G(2)-M Commun. 284, 880 886.
arrest. Cell Growth Dier. 13, 237 246.

Вам также может понравиться