You are on page 1of 18

Biochem. J.

(2007) 403, 217234 (Printed in Great Britain)

217

doi:10.1042/BJ20070024

REVIEW ARTICLE

Signalling to translation: how signal transduction pathways control the


protein synthetic machinery
Christopher G. PROUD1
Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3

Recent advances in our understanding of both the regulation


of components of the translational machinery and the upstream
signalling pathways that modulate them have provided important
new insights into the mechanisms by which hormones, growth
factors, nutrients and cellular energy status control protein
synthesis in mammalian cells. The importance of proper control of
mRNA translation is strikingly illustrated by the fact that defects
in this process or its control are implicated in a number of disease
states, such as cancer, tissue hypertrophy and neurodegeneration.
Signalling pathways such as those involving mTOR (mammalian
target of rapamycin) and mitogen-activated protein kinases modulate the phosphorylation of translation factors, the activities of the

protein kinases that act upon them and the association of RNAbinding proteins with specific mRNAs. These effects contribute
both to the overall control of protein synthesis (which is linked to
cell growth) and to the modulation of the translation or stability of
specific mRNAs. However, important questions remain about both
the contributions of individual regulatory events to the control
of general protein synthesis and the mechanisms by which the
translation of specific mRNAs is controlled.

INTRODUCTION

Conversely, and fourthly, protein synthesis places heavy demands


upon the cell in terms of requirements for both (essential) amino
acids as precursors and metabolic energy, because protein synthesis consumes a high proportion of cellular ATP and GTP. In
view of the above considerations, it is clear that overall protein
synthesis must be subject to tight control to match the requirements of the cell/tissue with the supply of metabolic energy and
amino acids.
These regulatory mechanisms primarily involve rapid (minutes)
changes in the activity or association of components of the translational machinery, which are primarily mediated by changes
in the states of phosphorylation, e.g. of translation factors and
specific RNA-binding proteins (see Figure 1 for a simplified
overview). Over the longer term (hours to days), the control of protein synthesis involves changes in the cellular levels of translation
factors and ribosomes, i.e. in the cells capacity for protein synthesis. The pathways that control translational capacity mesh with
those that control of translation factor activity, presumably to
ensure co-ordinated control of protein synthesis.
Recently, there have been substantial advances in our understanding of both the control of the translational apparatus and the
signal transduction pathways involved. The aim of this article is
to review these topics and to highlight some key questions that

Regulating the overall rate of protein synthesis is important for


modulating cell and tissue metabolism. One important example
of this is the anabolic effect of insulin is to activate protein synthesis in tissues such as fat and skeletal muscle. Secondly, cell
and tissue growth require enhanced rates of protein synthesis.
On the other hand, excessive protein accumulation may lead to
tissue hypertrophy, e.g. in the case of cardiac muscle [1] and certain types of benign tumours (such as hamartomas) [2]. Cell proliferation also depends upon maintaining an adequate rate of
protein synthesis to allow cells to become large enough to generate
two daughter cells, although the links between mammalian cell
size and division are not clear-cut (see, e.g., [3]). Thirdly, control of the translational machinery also plays key roles in controlling gene expression. It is therefore not surprising that specific
translation factors and the signalling pathways that control them
are strongly implicated in the dysregulation of cell function that
leads to transformation and cancer. In particular, in many cases,
the synthesis of specific proteins involves or depends upon tight
control over the translation of their mRNAs. There are now
numerous examples of this kind of qualitative control of protein
synthesis, a few selected examples of which are discussed here.

Key words: mammalian target of rapamycin (mTOR), mRNA,


mRNA translation, ribosome, signal transduction, translation
factor.

Abbreviations used: AMPK, AMP-activated protein kinase; ARE, AU-rich element; ARE-BP, ARE-binding protein; ATM, ataxia telangiectasia mutated;
CaM, calmodulin; CDK, cyclin-dependent kinase; DYRK, dual-specificity tyrosine-phosphorylated and -regulated kinase; eEF, eukaryotic elongation factor;
eIF, eukaryotic initiation factor; 4E-BP, eIF4E-binding protein; ERK, extracellular-signal-regulated kinase; FKBP12, FK506-binding protein 12; GAP, GTPaseactivating protein; GEF, guanine-nucleotide-exchange factor; GSK3, glycogen synthase kinase 3; hnRNP, heterogeneous nuclear ribonucleoprotein; HRI,
haem-regulated inhibitor; IRES, internal ribosome-entry site; IRS1, insulin receptor substrate 1; MAPK, mitogen-activated protein kinase; MEF, mouse
embryonic fibroblast; MEK, MAPK/ERK kinase; Met-tRNAi , initiator methionyl-tRNA; MK2, MAPK-activated protein kinase 2; Mnk, MAPK signal-integrating
kinase or MAPK-interacting kinase; mTOR, mammalian target of rapamycin; mTORC, mTOR complex; PABP, poly(A)-binding protein; PERK, protein kinase
R-like endoplasmic reticulum kinase; PI3K, phosphoinositide 3-kinase; PKA, protein kinase A; PKB, protein kinase B; PKC, protein kinase C; PKR, protein
kinase R; PP1, protein phosphatase 1; PTEN, phosphatase and tensin homologue deleted on chromosome 10; raptor, regulatory associated protein of
mTOR; rDNA, ribosomal DNA; siRNA, small interfering RNA; S6K, S6 kinase; 5 -TOP, 5 -tract of oligopyrimidines; TNF, tumour necrosis factor ; TOS,
target of rapamycin signalling; TSC, tuberous sclerosis complex; TTP, tristetraprolin; UTR, untranslated region; VWM, leucoencephalopathy with vanishing
white matter.
1
email cgpr@interchange.ubc.ca

c The Authors Journal compilation 
c 2007 Biochemical Society

218

C. G. Proud

will then outline the main signalling pathways that are known to
regulate them, and subsequently try to integrate these two strands
to provide an overall picture of how signalling pathways can
operate to modulate mRNA translation in mammalian cells.
eIF2/2B: recruitment of Met-tRNAi (initiator methionyl-tRNA)

The initiation stage of translation involves the recruitment of the


40 S ribosomal subunit to the mRNA and the identification of
the start codon. Initiation is a major site for control of mRNA
translation. Several major events are involved in this process,
including the recruitment of Met-tRNAi to the 40 S subunit
(Figures 2i and 2ii). This requires eIF2, a heterotrimeric GTPbinding protein which binds Met-tRNAi only in its GTP-bound
state. The GTP is hydrolysed during the initiation process and
eIF2 leaves the 40 S subunit as inactive eIF2 GDP. Regeneration
of active eIF2 GTP requires a GEF (guanine-nucleotide-exchange
factor) called eIF2B (Figure 3), a heteropentamer () [4].
eIF2B contains the catalytic domain towards its C-terminus
[6] and undergoes phosphorylation at a number of sites [7,8].
The extreme C-terminus of eIF2B binds eIF2 and contains two
phosphorylation sites for protein kinase CK2 that are required for
this interaction [7].
eIF2 is subject to regulatory phosphorylation (at Ser51 ;
Table 1). eIF2(P) acts as a competitive inhibitor of eIF2B, thus
blocking recycling of eIF2 and inhibiting translation initiation
(Figure 3). Four eIF2 kinases are known in mammalian cells.
They share strong similarity in their catalytic domains, but possess
distinct regulatory domains which confer control generally under
stressful conditions [9]. As these kinases are not known to be
regulated by signalling pathways, they will not be discussed in
detail here (for recent reviews, see [9,10]).
The fact that all four kinases ultimately control the activity of
eIF2B suggests that regulating eIF2B is of key importance for the
control of translation. This is borne out by the findings: (i) that
overexpression of the catalytic subunit of eIF2B () leads to the
growth of neonatal cardiomyocytes [11], a process that is driven
by increased rates of protein synthesis [1], and (ii) that eIF2B
is activated by a diverse range of stimuli that turn on mRNA
translation, including insulin and growth factors [1214]. Such
activation occurs without discernible decreases in the phosphorylation of eIF2, indicating that additional mechanisms control
eIF2B activity. As mentioned above, eIF2B is a phosphoprotein
[7,15], and phosphorylation modulates its activity [see also under
PI3K (phosphoinositide 3-kinase) below] (Figure 3).
An interesting feature of eIF2B is that the severe inherited
neurodegenerative disease VWM (leucoencephalopathy with vanishing white matter), a demyelinating disorder also termed childhood ataxia with CNS (central nervous system) hypomyelination,
is caused by mutations in the genes for eIF2B [16,17]. Mutations
in any of the five genes (; also termed EIF2B1EIF2B5) can
cause VWM [18]. The phenotypes observed in VWM are highly
variable in terms of severity, age of onset and the range of lesions
observed [17,18]. Although it is now clear that VWM mutations
impair eIF2B function in a variety of ways [19,20], the actual basis
of the disease is unclear. These findings emphasize the crucial
importance of the normal function or control of eIF2B for normal
cell physiology.

Figure 1 Overview of the control of protein synthesis by signal transduction


pathways
Signalling pathways activated by receptors at the plasma membrane regulate translation factors
required for general translation and thereby modulate overall protein synthesis. They may also
regulate certain translation factors and mRNA-specific binding proteins to alter the rates of
translation (or the stability) of specific mRNAs and thus control translation in an mRNA-specific
manner. The availabilities of metabolic energy and of amino acids also provide important inputs
to the signalling processes that control mRNA translation. This depiction is purely schematic
and not exhaustive. A more detailed scheme is presented in Figure 8.

remain to be answered. A complete picture of the control of protein


synthesis is critical for our understanding of the regulation of cell
growth and proliferation.
OVERVIEW OF THE TRANSLATIONAL MACHINERY

The process of protein synthesis or mRNA translation is conventionally divided into three main stages: initiation, elongation and
termination. Each stage requires translation factors that transiently
associate with the ribosome. This review is concerned exclusively
with the regulation of translation in mammalian cells, where these
factors are termed eukaryotic initiation, elongation and termination (release) factors (eIFs, eEFs and eRFs). The purpose of this
review is not to describe in detail the functions of individual
translation factors (for reviews on this subject, see, e.g., [4,5]), but
rather to describe how intracellular signalling pathways control
them to modulate specific steps in mRNA translation.
This article thus focuses primarily on the translation factors
that are known to be subject to direct regulation, and a number of
aspects of these proteins are summarized in Table 1. In structuring
this article, I will start by providing an overview of the roles of the
relevant translation factors and other translational components. I

c The Authors Journal compilation 
c 2007 Biochemical Society

Recruitment of the ribosome to the mRNA

eIF4E binds the 5 -cap structure of the mRNA, which includes a 7methylguanosine moiety (Figure 2i). eIF4E contains a single site
of phosphorylation (Ser209 in mammals; Table 1). Early work suggested that phosphorylation of eIF4E enhanced its binding to cap

Signal transduction pathways and the control of protein synthesis


Table 1

219

Regulatory phosphorylation events that impinge upon mammalian translation factors and related proteins

Listed are phosphorylation sites in components of the mammalian translational machinery that are known to be regulated and/or to affect the activity of the corresponding proteins. The Table is not
intended to be an exhaustive list of all the phosphorylation that have identified in translation factors.
Protein

Phosphorylation site(s)

Kinase(s) responsible

Comments

Reference(s)

eEF1A

Thr431

eEF1B*

: Ser106 , Ser112

PKC

Activation?

Reviewed in [28,206]

: Thr230

CK2
PKC
CK2
PKC
CDK1

Effect unclear
Activation?
Effect unclear
Activation?
Inhibition?

eEF2

Thr56

eEF2 kinase

Inhibits ribosome binding

eIF2

: Ser51

HRI, PKR, mGCN2, PERK

Inhibits eIF2B-mediated recycling

[208]

eIF2B

: Ser540
Ser544
Ser717 /Ser718

GSK3
DYRK group
CK2

Inhibits recycling of eIF2


Primes GSK3
Allows eIF2 binding

[7,15]

eIF4B

Ser322

S6K

Facilitates binding to eIF3

[49,51]

eIF4E

Ser209

Mnk1/2

Decreases binding to 5 -cap structure

[209]

eIF4G

Ser1108 , Ser1148 , Ser1192


Not known

mTOR?
Mnks?

Functional effects not known

[210]
[173]

S6

Ser235 , Ser236 , Ser240 , Ser244 , Ser247

S6K1/2

Knock-in mouse created, but function remains unclear

[211]; see also [47]

eEF2 kinase

Ser78
Ser359
Ser366
Ser398
Ser500

?
?
S6K1, p90RSK
AMPK
PKA

Inhibits CaM binding


Inhibition
Inhibition
Activation?
Elicits Ca2+ -independent activity

[134]
[136]
[50]
[154]
[38]

: ?

mTOR
Priming
sites
Thr37 , Thr46

Ser64
mTOR

Thr70
Interferes with binding to eIF4E; unclear
mTOR?/CDK1?

CDK?
Ser112
* The different naming systems for subunits of eEF1B are complex and can be confusing. The system adopted here is that proposed by Merrick and Nyborg [215].
The -subunit of eIF2 is also a phosphoprotein, but the regulatory significance of this is unclear.
Other sites are known, but, as their functions are not entirely clear, they are not listed here.

4E-BP1

analogues or capped RNA [21]. Later reports, using fully defined


components, showed that phosphorylation actually decreases its
affinity for 7-methylguanosine or capped RNA [22,23].
eIF4E also binds a number of protein partners including eIF4G,
a multidomain scaffold protein which interacts with other components of the translational machinery (Figures 2 and 4A). These
include eIF4A, an RNA helicase that is thought to unwind regions
of secondary structure in the 5 -UTRs (untranslated regions) of
mRNAs. As such features can inhibit normal cap-dependent translation initiation (depicted in Figure 2iii), increased binding of
eIF4A to eIF4G/eIF4E should stimulate this process, especially
for mRNAs whose 5 -UTRs contain significant secondary structure. The helicase function of eIF4A is enhanced by eIF4B, which
interacts with both eIF4A and eIF3 [24]. eIF4B and eIF4G are both
phosphoproteins, as discussed below. The eIF4AeIF4EeIF4G
complex is often referred to as eIF4F [24] (Figure 4A).
The N-terminus of eIF4G binds the PABP [poly(A)-binding
protein], leading to circularization of the mRNA and enhancement of its translation. eIF4G also binds to eIF3 [26]. Mammalian
eIF3 possesses 12 subunits and binds to the ribosomes 40 S subunit. It thus provides a key link between the mRNA (which binds
via eIF4E, PABP and other interactions to eIF4G and its partners)
and the 40S subunit.
The biological activity of eIF4E can be regulated by a set of 4EBPs (eIF4E-binding proteins). Three 4E-BPs exist in mammals,
although only 4E-BP1 has been studied in much detail. The 4EBPs interact with eIF4E [27] via a binding motif which resembles
those found in eIF4GI and eIF4GII , i.e. with the sequence

[207]

[212]
[213,214]

XXXX, where  is a hydrophobic residue and X is any


amino acid. By binding eIF4E, the 4E-BPs block its interaction
with eIF4G, thereby making eIF4E unavailable for initiation complex formation [24,27]. The 4E-BPs are phosphoproteins. 4E-BP1
contains at least seven sites of phosphorylation (Figure 4B and
Table 1), of which four are known to be regulated via signalling
pathways (Thr37 , Thr46 , Ser65 and Thr70 in human 4E-BP1).
Elongation factors

In mammalian cells, peptide-chain elongation requires two eEFs.


eEF1 binds GTP and recruits aminoacyl-tRNAs to the A-site of
the ribosome to match the codon located in that site. It is actually
eEF1A that does this, whereas eEF1B (a heterotrimer) acts as the
GEF for eEF1A, as eIF2B does for eIF2. eEF1A and subunits of
eEF1B are targets for phosphorylation by several kinases [28].
These include PKA (protein kinase A), PKC (protein kinase C)
and protein kinases involved in cell cycle control [notably CDK1
(cyclin-dependent kinase 1)/cyclin B], as described in detail in
[28] (Table 1). It remains to be established what effects these
modifications have on elongation rates, although there is evidence
that elongation is controlled during mitosis and that eEF1B localization changes during the cell cycle [28].
eEF2 is a monomer that binds GTP. It mediates the translocation step of elongation where the ribosome moves by one codon
relative to the mRNA and the peptidyl-tRNA moves from the Ato the P-site [29]. eEF2 is a phosphoprotein. In vivo, phosphorylation occurs at a single site (Thr56 [30]) and this impairs the

c The Authors Journal compilation 
c 2007 Biochemical Society

220

C. G. Proud

Figure 3

Targets for PI3K signalling include eIF2B and TSC2

eIF2B catalyses GDP/GTP exchange on eIF2, a heterotrimer of which the -subunit is the
guanine-nucleotide-binding component. eIF2B is a heteropentamer (). For simplicity, only
the catalytic, , subunit is depicted here. eIF2B is a phosphoprotein. Only two of the six known
sites are indicated: the GSK3 site (phosphorylation of which inhibits the activity of eIF2B) and
the priming site required by GSK3, which is phosphorylated by members of the DYRK family,
at least in vitro . Residue numbering for eIF2B is based on the human protein. TSC2 function
appears to be inhibited following its phosphorylation by PKB, and may be activated by GSK3.
See also Figure 5.

Figure 2

Initiation pathway for mRNA translation

This scheme only indicates the major steps, i.e. (i) binding of eIF4F and other factors to the cap,
mediated by eIF4E and its scaffold partner, eIF4G, which also binds the eIF4E kinases, the Mnks,
(ii) recruitment of Met-tRNAi , mediated by eIF2, (iii) scanning and subsequent start codon
recognition, which is followed (iv) by release of initiation factors, (v) the addition of the 60 S
subunit, and (vi) the commencement of elongation. This Figure is not intended to be detailed or
complete. m7 GTP, 7-methylguanosine triphosphate. To see an animated version of this Figure,
go to http://www.BiochemJ.org/bj/403/0217/bj4030217add.htm.

interaction of eEF2 with the ribosome, thus inactivating it [31].


Phosphorylation is catalysed by an unusual and highly specific
enzyme, eEF2 kinase (Figure 4C). Its catalytic domain shows
no sequence similarity to the main protein kinase superfamily
and it belongs to a distinct small family of protein kinases
have been termed -kinases [32,33]. The three-dimensional
structure of one member of this family [the cytoplasmic domain
of the TRP (transient receptor potential) channel Chak1] has been
solved crystallographically [34]. Despite the lack of sequence
similarity between Chak1 and other protein kinases, the threedimensional structure revealed that the binding between Chak1
and the nucleotide substrate probably includes similar interactions
to those seen in conventional protein kinases.
eEF2 kinase is normally completely dependent upon Ca2+ and
CaM (calmodulin) for activity (reviewed in [35]). The CaMbinding site is situated immediately N-terminal to the catalytic domain. The C-terminal region of eEF2 kinase contains several phosphorylation sites, as discussed below. Only one will be dealt with
in this section, as it is phosphorylated by PKA (Ser500 in human
eEF2 kinase [36,37]; Figure 4C), which will not be discussed

c The Authors Journal compilation 
c 2007 Biochemical Society

anywhere else in this review. Phosphorylation of eEF2 kinase at


Ser500 leads to acquisition of Ca2+ -independent activity [36,37].
At resting levels of Ca2+ , this is likely to correspond to kinase
activation, leading to increased phosphorylation and inactivation
of eEF2 [38,39]. Bearing in mind that the overwhelming majority
of the energy used in translation (> 99 %) is consumed during
elongation, it is tempting to speculate that this may allow agents
that activate PKA to slow down elongation and conserve energy
which can be diverted for other requirements, e.g. in adipocytes
stimulated by -adrenergic agonists [38].
The ability of Ca2+ ions to activate eEF2 kinase and inhibit
eEF2 may allow neural signals to inhibit the rate of elongation to
decrease energy utilization in contracting muscle and to complement their ability to turn on contraction, which requires energy.
The activation of eEF2 kinase by Ca2+ is also proposed to play a
role in reprogramming translation in neuronal cells in response
to neurotransmitter stimulation [40,41]. It is argued that, by slowing elongation and making this, rather than initiation, limiting for
translation, mRNAs that normally initiate translation inefficiently
would be more easily able to enter polysomes and be translated.
Similar effects are seen with cycloheximide, a reagent that blocks
elongation [42]. Other neuromodulatory stimuli such as 5-hydroxytryptamine (serotonin) and BDNF (brain-derived neurotrophic
factor) decrease eEF2 phosphorylation [43,44]. Given the growing
body of evidence that controlling translation is important in
learning, memory and behaviour [45,46], it will be important
to study the role of regulating elongation in these processes.
Ribosomal protein phosphorylation

Certain ribosomal proteins are also phosphorylated within cells.


The best-known of these is S6, a component of the 40 S subunit.
In mammals, there are two genes for S6Ks (S6 kinases), S6K1 and
S6K2 (Figure 4D). Each gives rise to two transcripts and two distinct polypeptides, which differ at their N-termini by a sequence
containing a potential nuclear localization signal [47]. The activation of the S6Ks involves their phosphorylation at multiple sites
(Figure 4D and Table 1).
In addition to S6, which is discussed in detail below, other substrates for the S6Ks have also been identified. Within the realm of

Signal transduction pathways and the control of protein synthesis

221

Regulation of the translation of specific mRNAs

There are far too many examples of the translational control of


specific mRNAs to provide comprehensive coverage here. Such
control allows cells rapidly to adjust the rate of synthesis of specific proteins in response to changing conditions. Enhancing the
translation of a specific mRNA rapidly steps up production of
the corresponding protein, without the need to turn on transcription, process the mRNA and export it to the cytoplasm. There are,
moreover, many examples of translational control being exploited
to achieve spatial or temporal control of protein expression in early
development [52]. Within this review, only two classes of mRNAs
that are subject to specific translational control will be discussed:
i.e. the 5 -TOP (5 -tract of oligopyrimidines) mRNAs and mRNAs
containing AREs (AU-rich elements) in their 3 -UTRs.
UPSTREAM SIGNALLING TO THE TRANSLATIONAL MACHINERY
PI3K

The lipid kinase PI3K plays a key role in signalling downstream


of a wide variety of receptors, including those for insulin and the
insulin-like growth factors. Activation of PI3K leads to increased
production of phosphatidylinositol 3,4-bisphosphate, which activates effectors such as PKB (protein kinase B, also termed Akt)
[53] (Figure 3). The action of PI3K is opposed by the lipid phosphatase PTEN (phosphatase and tensin homologue deleted on
chromosome 10), a tumour suppressor. Mutations in PTEN occur
in many human tumours [54], leading to constitutive activation of
PKB and, consequently, of mTOR signalling (see below).
Regulation of eIF2B via PKB and GSK3 (glycogen synthase
kinase 3)
Figure 4

Targets for mTOR signalling

(A) Control of the eIF4F initiation factor complex. Binding of 4E-BPs (e.g. 4E-BP1) to eIF4E
occludes their binding site for eIF4E, thus preventing the formation of productive eIF4F initiation
factor complexes. mTOR-regulated phosphorylation of 4E-BP1, which occurs at multiple sites
(see the text and B) results in the release of 4E-BP1 from eIF4E, allowing eIF4G to bind and recruit
other translation factors, such as the helicase eIF4A, to the 5 -end of the mRNA. eIF4G also binds
PABP and the eIF4E kinases, the Mnks. (B) 4E-BP1 undergoes phosphorylation at multiple sites
in vivo . At least four of them are regulated by mTORC1 in response to amino acids and/or stimuli
such as insulin. The N-terminal sites depend on the N-terminal RAIP motif, whereas Ser65 and
Thr70 depend both on the phosphorylation of these sites and on the TOS motif. These latter
sites are adjacent to the eIF4E-binding motif and control binding to eIF4E (see the text), which
occurs through the indicated consensus motif. Less is known about the function or regulation
of the other sites. (C) eEF2 kinase. The CaM-binding domain and the unusual catalytic (kinase)
domain are shown, as well as the C-terminal region that is required for efficient phosphorylation
of eEF2. eEF2 kinase undergoes phosphorylation in vivo at multiple sites, a selection of which
is shown. Where known, the effect of these phosphorylation events on eEF2 kinase activity is
indicated (orange circles, inhibition; green hexagons, activation) as is the identity of the kinase
responsible. In some cases (???), the corresponding kinase is not known or additional kinases to
that shown are involved. (D) S6Ks: the overall structural organiziation of S6K 1 and 2 is similar.
Each exists as two splice variants, the longer of which contains an N-terminal extension
that includes a nuclear- or nucleolar-targeting signal. C-terminal to the kinase domain lie
a number of sites whose phosphorylation is essential for full kinase activity: one of these
(Thr412 ) can be phosphorylated by mTOR in vitro . Residue numbering is based upon the longer
form of human S6K1, since S6K1 has been studied in much more detail than S6K2. Almost
all of the regulatory phosphorylation sites identified in S6K1 are conserved in S6K2.

mRNA translation, these include eIF4B [48,49] and eEF2 kinase


[50]. In both cases, the effect of S6K-mediated phosphorylation
favours activation of translation: for eIF4B, it increases its association with eIF3 [48,51] and it causes inactivation of eEF2 kinase
[50], providing a link between mTOR (mammalian target of
rapamycin; see below) and the control of eEF2.

One substrate for PKB is GSK3, which is named for its role
in controlling the rate-limiting enzyme of glycogen deposition,
but which also plays diverse roles in other important cellular processes, including development, gene transcription and the organization of the microtubular cytoskeleton [55,56]. PKB phosphorylates both GSK3 isoforms ( and ) at their N-termini, inhibiting
their activity against certain substrates, including eIF2B [8,55]
(Ser540 in human eIF2B; Figure 3 and Table 1). Such substrates
are ones whose phosphorylation by GSK3 requires a priming
phosphorylation event at a serine residue four residues C-terminal
to the GSK3 target site. In the case of eIF2B, the priming kinases
may be members of the DYRK (dual-specificity tyrosine-phosphorylated and -regulated kinase) group of enzymes [57] (Figure 3). Both the GSK3 and priming sites are found in Drosophila
eIF2B, but not in the orthologues from yeast [58]. The phosphorylation of eIF2B by GSK3 inhibits its activity [15]. The
inactivation of GSK3 by insulin thus leads to dephosphorylation
of the inhibitory GSK3 site on eIF2B and to activation of eIF2B.
This provides a mechanism whereby agents such as insulin can
activate eIF2B, a key component for general translation (Figure 3).
It has also been reported that eIF2B may play a role in the anti- and
pro-apoptotic effects of signalling through PKB/Akt and GSK3
respectively [59]. However, the mechanisms remain unclear; it
is tempting to speculate that inhibition of eIF2B may promote
the translation of proteins that play a pro-apoptotic role (or
vice versa). Inhibition of eIF2B activity via phosphorylation of
eIF2 does lead to the specific up-regulation of the translation
of certain mRNAs, such as those for GCN4 (general control nonderepressible 4) in yeast and for ATF4 (activating transcription
factor 4) in mammals [4,60].

c The Authors Journal compilation 
c 2007 Biochemical Society

222

C. G. Proud

Although, such as by insulin, activating PKB and inhibiting


GSK3 elicits the dephosphorylation of the GSK3 site in eIF2B,
the dephosphorylation of this site seems to be insufficient, by
itself, to evoke activation of eIF2B [61]. This implies that other
regulatory inputs may be important. There are reports of other
inputs to eIF2B from amino acids [62] and from ERK (extracellular-signal-regulated kinase) signalling [63,64]. The input from
ERK to the control of eIF2B appears to be mediated via activation
of PP1 (protein phosphatase 1), although it is not known which
phosphorylation site in eIF2B is involved [64,65]. Further work
is necessary both to determine the molecular basis of these inputs
and to define the importance of the control of eIF2B for the overall
activation of protein synthesis.
The N-terminal sites in GSK3 can also be phosphorylated by
p90RSK which lies downstream of ERK signalling (see below).
Although this could provide a mechanism by which Ras/ERK signalling could activate eIF2B, the available data suggest that the
activation of eIF2B via this pathway does not involve changes in
the phosphorylation of the GSK3 site in eIF2B [63] and seems,
instead, to be mediated by changes in the activity of a form of
PP1 [65].
mTOR, a master regulator of the translational machinery

mTOR plays a key role in regulating the phosphorylation of a


number of components of the translational machinery. mTOR is
a multidomain protein which interacts with a number of other
proteins (Figure 5). Its N-terminus contains several HEAT
(Huntingtin, elongation factor 3, the PR65/A subunit of protein
phosphatase 2A and the lipid kinase Tor) domains (modules for
proteinprotein interactions) and its C-terminus includes a kinase
domain similar to the PI3Ks, and thus to other PI3K-related
kinases, such as ATM (ataxia telangiectasia mutated), ATR (ATMand Rad3-related) and the DNA-activated protein kinase [66].
Immediately N-terminal to its kinase domain is a region that binds
the immunophilin FKBP12 (FK506-binding protein 12) when that
protein is bound to rapamycin [67]. Rapamycin/FKBP12 inhibits
some, but by no means all, of the functions of mTOR.
In vitro, mTOR exhibits protein kinase activity. This activity
is higher in the presence of high (non-physiological) concentrations of Mn2+ than in the presence of physiological levels of
Mg2+ . In vitro, mTOR can phosphorylate proteins such as 4E-BP1
and S6K1 at sites that are phosphorylated in an mTOR-dependent
manner in vivo [68,69]. Interestingly, the phosphorylation of
Thr37 / Thr46 in 4E-BP1 is relatively insensitive to rapamycin in
living cells [70] and is also only partially (< 30 %) inhibited by
rapamycin in vitro (in kinase reactions using immunoprecipitated
mTOR) [71].
The phosphorylation of 4E-BP1 and S6K1 is stimulated by
serum, insulin and growth factors. In many cell types, this requires
that cells are provided with amino acids; amino acid starvation
causes a swift impairment of mTOR signalling, as indicated by dephosphorylation of 4E-BP1 and S6K1, and agents such as insulin
can no longer (fully) induce their phosphorylation. The most
effective single amino acid in most cases is leucine, an essential
branched-chain amino acid [72]. It is likely that leucine acts within
the cell and it seems that its metabolism is not required. However,
it is not yet clear how it activates mTOR signalling (Figure 5).
mTOR forms two types of multisubunit complex

A major advance in recent years has been the discovery that mTOR
(and its orthologues in species as evolutionarily distant as budding
yeast) form at least two types of complex involving distinct partner
proteins (Figure 5). This topic has been the subject of several

c The Authors Journal compilation 
c 2007 Biochemical Society

recent articles [67,73], and will not be dealt with in detail here.
The key point is that at least two distinct kinds of complex exist in
mammals. One complex, mTORC (mTOR complex) 1 contains
the partner raptor (regulatory associated protein of mTOR; termed
KOG1 in yeast); the second one, mTORC2, contains a different
partner, rictor (rapamycin-insensitive companion of mTOR;
mAVO3). Both mTORC1 and mTORC2 contain the protein GL
(also termed mLst8). mTORC1 mediates effects of mTOR that
are sensitive to rapamycin, whereas effects mediated through
mTORC2 are insensitive to this compound. The reasons for this
difference, and indeed the mechanisms by which rapamycin impairs functions of mTORC1, are still not completely clear. Some
studies show that treatment of cells with rapamycin destabilizes
the mTORraptor (mTORC1) complex (see, e.g., [74]), but other
investigators have found that raptor could be purified with mTOR
as a complex with FKBP12/rapamycin [75], which runs contrary
to the idea that rapamycin causes dissociation of this complex.
Raptor interacts with the TOS (target of rapamycin signalling)
motifs in targets of mTOR that are regulated in a rapamycinsensitive manner. This presence of the TOS motif and thus, one
assumes, its binding to raptor enhances the ability of mTORC1
to phosphorylate proteins 4E-BP1 and S6K1 in vitro [76,77].
Indeed, the presence of raptor in mTOR immunoprecipitates
greatly enhances the phosphorylation of 4E-BP1 by mTOR
in vitro [76].
Effects due to mTORC2 include the phosphorylation of the
hydrophobic motif site in PKB [7880] [which is equivalent to
the mTORC1 site (Thr389 ) in S6K1]. Since it is mTORC1, not
mTORC2, that is linked to the control of components of the translational machinery, the remaining discussion of mTOR will focus
on mTORC1.
Upstream control of mTORC1

How do agents such as insulin activate mTORC1 signalling?


The generally accepted model for this is depicted in Figures 3
and 4. By activating PKB, insulin elicits the phosphorylation of
TSC2 (tuberous sclerosis complex 2), one component of a dimeric
complex also containing TSC1 (Figure 5). TSC2 acts as a GAP
(GTPase-activating protein) for the small G-protein Rheb [81,82].
Rheb interacts with mTOR [via the N-terminal (ATP-binding
region) of its kinase domain]. Unusually for a G-protein, Rheb
interacts with mTOR irrespective of its guanine-nucleotide-binding status [83,84]; however, only Rheb GTP was found to stimulate the in vitro kinase activity of mTOR [83]. These findings
have led to the concept (Figures 3 and 5) whereby the PKBmediated phosphorylation of TSC2 leads to the inhibition of its
GAP activity towards Rheb, allowing Rheb to accumulate in
its GTP-bound state, leading to the activation of mTORC1 (Figure 5). However, there is a lack of direct evidence that phosphorylation of TSC2 inhibits its GAP activity. Alternatively,
phosphorylation of TSC2 may alter its stability, its association
with TSC1 or its intracellular localization [85,86].
As described above, loss of PTEN function causes activation of
PKB and thus stimulation of mTORC1 signalling. Interestingly,
the proliferation of some tumour-derived cell lines (e.g. ones lacking the tumour suppressor PTEN) is extremely sensitive to inhibition by rapamycin, implying a key role for signalling through
mTORC1 [87]. Thus activation of mTORC1 resulting from constitutive PKB activation leads to a transformed phenotype. This
contrasts with the effect of loss of TSC1TSC2 function, which
also results in activation of mTOR, but (usually) leads to nonmalignant tumours characterized by very large cell size, termed
hamartomas [88]. This difference probably reflects additional
roles that PKB/Akt plays in cell survival and other processes [89].

Signal transduction pathways and the control of protein synthesis

Figure 5

223

mTOR complexes

The mTORC1 and mTORC2 complexes are shown schematically. Some other proteins have been reported to bind mTOR, but are not shown here for simplicity. mTORC2 phosphorylates PKB, but
little is known about its regulation. mTORC1 phosphorylates S6Ks and 4E-BPs in vitro (although additional kinases may be involved in their phosphorylation in vivo ). The kinase activity of mTOR
has been shown to be stimulated, in vitro , by Rheb GTP. The hydrolysis of Rheb-bound GTP to GDP is stimulated by TSC2, which, together with TSC1, negatively regulates mTORC1. TSC2 can be
phosphorylated at multiple sites by the indicated kinases. The functional effects of these phosphorylation events remain to be clarified, but they are presumed to activate (green hexagons) or inhibit
(orange circles) TSC2 function. An unfilled circle indicates uncertainty about the direction of the effect. Residue numbers are based on human TSC2; owing to the existence of distinct splice variants,
numbering systems vary, so that numbers used here do not correspond to those used by certain authors. Signalling through mTORC1 is also activated by leucine, by an unknown mechanism. This
does not require TSC2.

Activation of mTORC1 is also linked to a range of other oncogenes or proto-oncogenes, including PI3K, Akt, Ras, NF (nuclear
factor) and LKB1 (reviewed in [87,9092]).
In amino-acid-starved cells, the overexpression of Rheb restores mTOR signalling (see, e.g., [93]). This gave rise to the idea
that amino acids may, similarly to insulin, enhance the proportion
of Rheb in its GTP-bound state, perhaps by interfering with TSC2
function. However, amino acid starvation has, at most, only modest effects on the proportion of Rheb bound to GTP [84,94], and
amino acid starvation still impairs mTOR signalling in TSC2null cells [84]. This indicates that amino acids can work in a
TSC2-independent way to regulate mTOR. The results presented
by Long et al. [83] indicate that amino acid starvation somehow
impairs the interaction between Rheb and mTOR. It is an important priority to discover how amino acids do this.

mTORC1 regulates the phosphorylation of 4E-BP1 and S6K1


at multiple sites

The 4E-BPs and S6Ks each contain a TOS motif which binds the
mTORC1 component, raptor. Each of these proteins is subject to
phosphorylation at multiple sites, many of which undergo dephosphorylation in response to starvation of cells for amino acids.
Phosphorylation of both proteins follows a hierarchy: in the case
of 4E-BP1, phosphorylation of Thr37 / Thr46 appears to be required
for modification of Thr70 , following which Ser65 undergoes phosphorylation; phosphorylation at Ser101 is also required for modification at Ser65 [9597]. Ser65 is generally the site that is mostly

strongly increased in response to agents such as insulin. Phosphorylation of Thr37 / Thr46 does not regulate the binding of 4EBP1 to eIF4E directly, whereas phosphorylation at Ser65 and Thr70
does, although their individual contributions to the control of
eIF4E binding is controversial (see, e.g., [70,97,98]).
Two short amino acid motifs play key roles in modulating 4EBP1 phosphorylation (Figure 4B). The TOS motif in the extreme
C-terminus is required for phosphorylation of Ser65 / Thr70 . In all
three mammalian 4E-BPs, the sequence of the motif is the same,
FEMDI (Phe-Glu-Met-Asp-Ile). The S6Ks (see below) also contain TOS motifs, their N-termini, with the related sequences
FDIDL (Phe-Asp-Ile-Asp-Leu) (S6K1) or FDLDL (Phe-AspLeu-Asp-Leu) (S6K2) [77,99]. The TOS motif is thought to bind
directly to the protein raptor, a partner of mTOR (see below also).
Phosphorylation of Thr37 / Thr46 does not require the TOS motif,
but depends instead on an N-terminal RAIP (Arg-Ala-Ile-Pro)
motif [70,100] (Figure 4B). Their phosphorylation is maintained
by amino acids (which activate mTOR signalling) and is affected
only modestly by agents such as insulin, which generally stimulates phosphorylation at Ser65 , and, more variably, Thr70 [70]. Phosphorylation of endogenous 4E-BP1 at Thr37 / Thr46 is also rather
insensitive to treatment of cells with rapamycin [70], but the evidence available strongly indicates that these sites are, nonetheless,
regulated by mTOR [70]. The relative rapamycin-insensitivity of
the N-terminal sites in 4E-BP1 (Thr37 / Thr46 ) could indicate that
are also targets for mTORC2. However, the available evidence
suggests this is not the case: 4E-BP1 is an extremely poor substrate
for phosphorylation by mTORC2 in vitro (B. D. Fonseca and
C. G. Proud, unpublished work).

c The Authors Journal compilation 
c 2007 Biochemical Society

224

C. G. Proud

The complex hierarchical phosphorylation of 4E-BP1 and its


dependence on two distinct motifs leads one to question whether
all the mTOR-regulated sites in 4E-BP1 and S6K1 are really direct
targets for phosphorylation by mTOR itself.
Other known targets of mTOR signalling, e.g. the S6Ks and
eEF2 kinase (see below), lack recognizable RAIP motifs. S6Ks
also undergo hierarchical phosphorylation at multiple sites, which
leads to their activation [47]. Interestingly, the main site that is
phosphorylated by mTOR in vitro, Thr389 , appears to be phosphorylated late in the hierarchy. It is the phosphorylation of this
site that correlates best with S6K activity [101]. However, the
phosphorylation of several other sites is also blocked by rapamycin, raising important questions about the identity of the
mTORC1-regulated mechanisms (kinases) that control these
modifications in vivo.
What are the physiological roles of the 4E-BPs and S6Ks? Findings
from 4E-BP-knockout mice

By binding to eIF4E, 4E-BPs prevent it from interacting with


the related scaffold proteins eIF4GI and eIF4GII [27,102] to form
initiation factor complexes competent to mediate cap-dependent
translation. However, their physiological roles in the normal
control of protein synthesis still remain to be fully established. At
most, only modest phenotypes have been reported for 4E-BP1knockout mice [103,104]. The latter paper reported alterations
in white and brown adipose tissue and metabolic rate, which
were associated with increased expression of PGC-1 (peroxisomeproliferator-activated receptor co-activator 1), a transcriptional
regulator involved in mitochondrial biogenesis and thus in
adaptive thermogenesis. Data from mice lacking 4E-BP2 indicate
a role for this protein in synaptic plasticity in the hippocampus;
such animals exhibit deficits in learning and memory [105].
Knocking out 4E-BPs affects the normal regulation of eIF4F
formation (e.g. knocking out 4E-BP2 increases basal eIF4F levels
[104]), implying that proper control of eIF4F is important for
normal cellular control, but the mechanisms involved remain
obscure. As many tissues express both 4E-BP1 and 4E-BP2 [104],
it will be important to study the phenotypes of animals in which
both have been knocked out, and ultimately to extend this to 4EBP3-knockout animals (see the Note added in proof at the
end of the Conclusions and perspectives section). The possible
mechanisms by which 4E-BP3 is controlled remain to be
investigated.
Links between eIF4E, 4E-BPs, mTOR and cancer

There is strong evidence that eIF4E plays an important role in cell


transformation and in human cancers [106,107]. For example,
overexpressing eIF4E in cell lines can cause their transformation.
eIF4E is expressed at high levels in many human cancers, levels
of expression correlating positively with the aggression of
the tumour. Transgenic animal models have confirmed a role
for eIF4E in transformation and tumour development [108,109].
eIF4E represses apoptosis [110,111] and targeted inhibition
of eIF4E induces rapid cell death [112]. How does eIF4E contribute to transformation and tumour progression? This may, of
course, involve its anti-apoptotic properties, but how does it exert
these effects? The most obvious way would be by promoting the
translation of the mRNAs for proteins with roles in cell-cycle
progression (e.g. cyclin D1 [113]), cell transformation (e.g. ornithine decarboxylase [114,115]), tumour vascularization {e.g.
VEGF (vascular endothelial growth factor) [116]} or metastasis
(e.g. matrix metalloproteinase-9 [117]) [107,118]. Such increased
translation could perhaps be due to the increased formation of

c The Authors Journal compilation 
c 2007 Biochemical Society

eIF4F complexes by out-titrating the inhibitory 4E-BPs (see


below). This is consistent with the ability of overexpression of
eIF4G to transform cells [119].
A further link to cell transformation may be the ability of eIF4E
to increase the cytoplasmic levels of the mRNA for cyclin D1, a
key regulator at the G1 /S boundary of the cell cycle [120]. This apparently involves a feature in the 3 -UTR of the cyclin D1 mRNA
[121], rather than the cap-binding function of eIF4E, which is
thought to allow eIF4E to promote transport of the cyclin D1
mRNA from the nucleus to the cytoplasm. Such a function would
require eIF4E to spend some time in the nucleus, and indeed a
proportion of the total cellular eIF4E is found in the nuclear fraction [122]. It remains to be established whether cyclin D1 mRNA
transport also involves eIF4E in cells where it is expressed at
normal levels. Priorities here are (i) to identify additional mRNAs
whose translation is up-regulated when eIF4E is overexpressed,
and (ii) to identify other messages whose nucleocytoplasmic
transport may be mediated by eIF4E. This will, one hopes, cast
much-needed new light on the mechanisms that underlie the role
of eIF4E in tumorigenesis.
There is currently a high level of interest in the role of mTOR
in cancer [87,91,107]. The fact that the availability of eIF4E is
controlled by signalling through mTOR provides one probable
link between mTOR signalling and tumorigenesis/cell proliferation.
The role of S6 phosphorylation remains unclear

The functional significance of the S6Ks is less clear. No clear


phenotypes related to translation have been reported for knockout
mice, even from mice lacking S6K1 and S6K2 [123]. The fact
that knocking out murine S6K1 [124] or its Drosophila orthologue
[125] leads to reduced animal size appears consistent with a role in
the positive control of protein synthesis and is consonant with the
effects of mutating the phosphorylation sites in mouse S6 [126].
Although S6 phosphorylation and the S6Ks were identified a
relatively long time ago, the physiological function of S6 phosphorylation remains obscure. It was thought previously to be important in controlling the translation of a specific set of mRNAs
(the 5 -TOP mRNAs). However, it is now clear that this is not
the case [123]. Meyuhas and collegues created knock-in mice and
cells in which all five regulated sites of phosphorylation within
S6 were altered to alanines (S6[5A]) [126]. These studies demonstrated that S6 phosphorylation is not required for control of 5 TOP mRNA translation. S6[5A] MEFs (mouse embryonic fibroblasts) actually showed higher rates of protein synthesis, but
were also smaller in size than wild-type MEFs [126]. Rapamycin
treatment decreased the size of wild-type MEFs, but did not affect
the size of S6[5A] MEFs [126], indicating that phosphorylation
of S6 is likely to play a role in cell size control, although how it
does so is unclear.
The major phenotypes reported in S6[5A] mice were decreased circulating insulin levels and enhanced peripheral insulin
sensitivity [126]. This appears to result from decreased overall
insulin production rather than reduced total -cell mass. S6[5A]
mice show relative glucose intolerance, whereas S6K1/ mice
show enhanced insulin sensitivity [127], apparently due to feedback inhibition by S6K1 at the level of IRS1 (insulin receptor
substrate 1)/PI3K [128,129]. The observation that S6[5A] mice
also show improved insulin sensitivity suggests that direct
phosphorylation of IRS1 by S6K1 is not the primary, or only,
mechanism involved here (see [126] for further discussion).
Cardiac hypertrophy involves increased cell (myocyte) size,
and is associated with elevated rates of protein synthesis [1].
Rapamycin inhibits, and even reverses, the development of cardiac

Signal transduction pathways and the control of protein synthesis

hypertrophy in vivo, indicating a key role for mTORC1 signalling


in this process [130,131]. Perhaps surprisingly, given the data
linking them to the control of cell size, knocking out S6K1 and
S6K2 in mice did not affect the development of physiological
(exercise-induced) or pathological hypertrophy [132].
eEF2 kinase is also regulated by mTOR

Insulin and a number of other agents rapidly induce the dephosphorylation of eEF2 [35]. The insulin-induced dephosphorylation
of eEF2 is associated with an accelerated rate of elongation [133].
Insulin causes the inactivation of eEF2 kinase and this involves a
decrease in its ability to bind CaM [133,134]. These effects are
blocked by rapamycin, identifying eEF2 kinase as a target for
regulation by mTOR. However, unlike the S6Ks and 4E-BPs,
eEF2 kinase does not contain an identifiable TOS motif, does not
interact with raptor and is not a substrate for phosphorylation by
mTOR in vitro (E. M. Smith and C. G. Proud, unpublished work).
This suggests that other signalling components act as intermediaries to link mTORC1 to the control of eEF2 kinase. Since the control of eEF2 has been the subject of other recent reviews [29,35],
discussion will focus on the links between mTORC1 and eEF2
kinase and on recent developments in understanding the control
of eEF2 kinase.
Three mTOR-regulated sites of phosphorylation have been
identified in eEF2 kinase (Ser78 , Ser359 and Ser366 ; Figure 4C).
Ser366 is phosphorylated by S6K1 and by the ERK-activated kinase
p90RSK [50,135], and this impairs the activity of eEF2 kinase at
low Ca2+ ion concentrations. Phosphorylation at Ser359
also inhibits eEF2 kinase activity (at saturating Ca2+ ion concentrations), and is regulated by amino acids and rapamycin
[134,136], suggesting that it is a target for an unidentified mTORregulated protein kinase. Most recently, phosphorylation at Ser78
has been shown to inhibit eEF2 kinase by interfering with its
ability to bind CaM [134]. This site lies immediately next to the
CaM-binding site in eEF2 kinase (Figure 4C). Its phosphorylation
is strongly promoted by insulin and blocked by rapamycin or
amino acid starvation. Again, it is not yet known which kinase
phosphorylates Ser78 .
Major goals is this area must be to determine the roles of these
three sites (and perhaps others) in the control of eEF2 kinase by
mTORC1 and amino acids; to identify the protein kinases that
phosphorylate Ser78 and Ser359 in eEF2 kinase, which may have
other roles in signalling downstream of mTOR; and to elucidate
the importance of the control of eEF2 to overall regulation of
protein synthesis under different conditions. An important step
toward the last goal is the production of animals and cells that are
devoid of eEF2 kinase activity [32].
Stress-activated kinase signalling also impinges on eEF2 kinase
[137]. SAPK4 (stress-activated protein kinase 4) [also called
p38 MAPK (mitogen-activated protein kinase) ] phosphorylates
Ser359 (which inactivates eEF2 kinase). Ser377 is a potential target
for several protein kinases, although phosphorylation here was
reported not to affect eEF2 kinase activity under the conditions
tested. It is possible that, like phosphorylation at Ser366 , it affects
calcium sensitivity, not maximal activity. Ser396 is a substrate for
several stress-activated kinases in vitro, and this phosphorylation
slightly inhibits eEF2 kinase activity.
5 -TOP mRNAs

The mRNAs encoding the cytoplasmic ribosomal proteins (rproteins) in mammalian cells are characterized by the presence
at their extreme 5 -end of a tract of pyrimidine nucleotides [138].
The translation of these mRNAs is subject to rapid regulation. In

225

serum-starved cells, these mRNAs (for example the mRNA for


eEF1A) are not associated with large polysomes and a proportion
actually runs in non-polysomal fractions on a sucrose density
gradient, whereas the remainder appears to be associated with
monosomes or disomes [139]. Following stimulation of cells
with serum, the mRNA (e.g. eEF1A) moves into large polysomes,
indicative of increased efficiency of translation of this message
[138,140]. This group of mRNAs also includes certain other
components of the translational machinery such as elongation
factors eEF1A and eEF2 [141], and PABP [142].
This shift (translational activation) is inhibited by rapamycin
[138,140], implying a role for mTORC1 signalling, although the
effect is only partial. Amino acid starvation appears to have a
greater inhibitory effect on the polysomal association of 5 -TOP
mRNAs (such as that for the 60 S subunit protein, L32) [143].
Since amino acids positively regulate mTOR, these data are
consistent with a role for mTORC1 in controlling 5 -TOP mRNA
translation (Figure 5).
The fact that 5 -TOP mRNA is sensitive to rapamycin had led
to the suggestion that their regulation might involve the S6Ks and
S6 phosphorylation. With the demise of this model [123,126], it is
now a high priority to establish how the translation of the 5 -TOP
mRNAs is controlled, and how mTORC1 signalling feeds into
this.
It is important to note that mTORC1 signalling also positively
regulates rDNA (ribosomal DNA) transcription (reviewed in
[144]), suggesting the possibility of co-ordinated regulation of
synthesis of the protein and RNA components of ribosomes, and
thus of ribosome biogenesis. A role for mTORC1 makes excellent
physiological sense, since mTORC1 co-ordinates multiple inputs
including those from amino acids (crucial for both ribosome biogenesis and mRNA translation), cellular energy and anabolic,
mitogenic and hypertrophic stimuli (all three of these require increased ribosome numbers to promote protein accumulation, cell
proliferation or cell growth). Fully elucidating the links between
mTORC1 and rDNA transcription/ribosome biogenesis is a major
goal in this area. This topic was recently discussed in detail [144].
Control of the translational machinery by cellular energy status

As alluded to already, protein synthesis is an energy-hungry process. The addition of each amino acid requires hydrolysis of the
equivalent of four ATP molecules. Estimates of the proportion of
the total cellular energy economy devoted to translation vary, and
of course this will differ a lot between different cell types, with
dividing cells presumably requiring more protein synthesis than
post-mitotic primary cells. A high proportion of cellular metabolic
energy is used in translation [145]. Almost all will be consumed in
the elongation phase. Actually making the components required
for translation, e.g. ribosomes, also requires substantial resources.
A number of connections between cellular energy status or
hypoxia, which leads to depletion of ATP, have been identified,
as recently reviewed in detail [146]. Dennis et al. [147] have
argued that the high K m of mTOR for ATP would make it able to
sense falling ATP levels, resulting in decreased mTOR kinase
activity and impairment of translation. However, such a mechanism could only come into play under very severe (non-physiological) conditions of energy depletion where ATP levels fall catastrophically. In contrast, AMPK (AMP-activated protein kinase) is a
much more sensitive system for reacting to changing adenine
nucleotide levels. AMPK is activated by AMP (normal 5 -AMP,
not cAMP) which is produced when ATP levels fall due to the
salvaging action of adenylate kinase: a small fall in ATP levels
actually causes a proportionally much larger increase in AMP,
so this energy-sensing mechanism is very sensitive to changes in

c The Authors Journal compilation 
c 2007 Biochemical Society

226

C. G. Proud

NTP levels. AMPK is a key component in the cellular energymanagement machinery [148]. It is therefore not surprising that
AMPK is linked to the control of the translational machinery,
although these links were only elucidated quite recently.
AMPK regulates mTORC1

AMPK phosphorylates TSC2 and turns off mTORC1 signalling,


presumably because AMPK-mediated phosphorylation of TSC2
stimulates its GAP activity towards Rheb [149]. Other potential
negative regulators of mTORC1 in response to hypoxia include
the stress-induced proteins RTP801 and RTP801L (also termed
Redd1 and Redd2), which inhibit mTORC1 signalling by acting
upstream of TSC1TSC2 [150,151].
Recent data point to convergence between Wnt signalling via
GSK3 and AMPK in the control of TSC2/mTORC1 [152,153].
Phosphorylation of TSC2 by AMPK provides the priming phosphorylation event that is required to allow GSK3 to phosphorylate
TSC2 at two sites. Mutation of these sites to residues that cannot
be phosphorylated sensitized cells to apoptosis induced by glucose (i.e. energy) deprivation. AMPK would thus provide an
essential input to allow control of TSC2 by GSK3. As described
above, insulin and other agents inhibit GSK3 via phosphorylation
mediated, for example, by PKB. It remains to be established what
role the inactivation of GSK3 by, for example, insulin plays in the
control of mTORC1 signalling alongside the direct phosphorylation of TSC2 by PKB. GSK3 is also inactivated by Wnt signalling, albeit by a different mechanism than its control by insulin.
Wnt signalling plays a key role in many cellular processes, including survival, proliferation and, above all, development. The recent
data show that Wnt signalling activates mTORC1 through GSK3
and TSC2. The input from GSK3 will require sufficient AMPK
activity to ensure that the priming site is phosphorylated. Given
the strong links between Wnt signalling and cancer, these data
suggest that intervening in mTORC1 signalling may be beneficial
in these types of cancers, in addition to others mentioned above.
AMPK also phosphorylates eEF2 kinase

A second link involves the direct phosphorylation of eEF2 kinase


by AMPK [154,155] at Ser398 (Figure 4C) which appears to activate eEF2 kinase. Conditions where AMPK is activated are associated with inhibition of elongation and increased phosphorylation
of eEF2 [155157]. Since most of the energy required for translation is used during chain elongation, it makes sense to slow
down this process when ATP is in short supply. This would also
preserve polysomes, allowing translation to resume quickly once
cellular energy status has improved. Inhibiting initiation, in contrast, would cause a loss of polysomes, making the resumption
of translation more complicated as ribosomes would have to be
recruited once more to the mRNA. The ability of AMPK, via
TSC2, to impair mTORC1 signalling [149,158] could also lead
to increased phosphorylation of eEF2, as mTORC1 negatively
regulates eEF2 kinase. The fact that treatment of TSC2-null cells
(TSC2/ MEFs) with agents that activate AMPK still causes
increased phosphorylation of eEF2 [84] indicates that the direct
control of eEF2 kinase by AMPK is important in controlling
the elongation machinery in the absence of a functional TSC2
RhebmTORC1 axis. Recent studies suggest that control of eEF2
by cellular energy status plays a role in shutting down protein
synthesis under conditions of hypoxia [158160], although other
mechanisms, including eIF2 phosphorylation, also contribute
to this [161,162]. Data from experiments where eEF2 kinase
expression was ablated by RNA interference indicate that it plays
an important role in the shut-off of protein synthesis in hypoxic

c The Authors Journal compilation 
c 2007 Biochemical Society

Figure 6

Links to MAPK signalling modules

(A) There are connections from ERK and p38 MAPK / signalling to the translational machinery
via the p90RSK group of kinases, which are activated by ERKs; MK2, which is activated by p38
MAPKs; and the Mnks, which are activated by both. MKKKs indicates that multiple upstream MAPK kinase kinases may activate MKK3 (MAPK kinase 3) and/or MKK6. (B) Schematic
illustration of the Mnks. The two human genes each generate two isoforms, yielding four
proteins which differ mainly at their C-termini. The N-termini of Mnk2a/b are longer than those
of Mnk1a/b. Major features are shown, including the threonine residues in the activation loop
which are phosphorylated by ERK/p38 MAPKs. The C-termini of Mnk1a and Mnk2a are similar
(but contain distinct MAPK-binding sites) and differ markedly from those of Mnk1b and Mnk2b.
Numbering is based on the human Mnks.

breast epithelial cells [159] and in the cytoprotective effect of


AMPK activation in neonatal cardiomyocytes [158].
MK2 (MAPK-activated protein kinase 2), a kinase that is
activated by p38 MAPK /, also phosphorylates TSC2 [163].
Since MK2 is switched on by cellular stresses and by cytokines,
this could provide an input to the control of mTOR, although this
remains to be clarified.
ERK and p38 MAPK signalling

Mammalian cells contain several MAPK signalling modules of


which the best understood are the classical MAPK (ERK), p38
MAPK / and JNK (c-Jun N-terminal kinase) pathways. There
are clear links between the first two and the control of the
translational machinery. Each involves downstream kinases that
phosphorylate components of the translational machinery and/or
other proteins that regulate mRNA translation (Figure 6A). ERK
activates members of the p90RSK group of kinases (RSK1RSK4)
which, as described below, phosphorylate several translation
factors or their regulators. p38 MAPK / activate MK-2, which

Signal transduction pathways and the control of protein synthesis

227

regulates the stability of mRNAs, such as that for TNF (tumour


necrosis factor ), probably via the phosphorylation of AREbinding proteins (see, e.g., [164167]). Detailed discussion of
the regulation of mRNA stability is outside the remit of the
present review. However, it is likely that controlling the translation
of ARE-containing mRNAs is also important for regulating the
expression of proteins that they encode. Recent data suggest
that the Mnks (MAPK signal-integrating kinases or MAPKinteracting kinases) may be important in this [168,169]. Controlling mRNA function at post-transcriptional levels, such as mRNA
stability or translation (rather than at transcription), allows much
faster responses. This is probably important under a wide range
of conditions, such as, in the case of TNF, combating infection.
Mnks

Mnk1 and 2 are activated by phosphorylation by either the classical MAPK, ERK1/2, or by p38 MAPKs or [170172]
(Figure 6A). They bind through a region in the N-termini to the
C-terminus of eIF4G and mediate eIF4E phosphorylation in vivo
[173,174].
In human cells, expression of each Mnk gene yields two different mRNAs by alternative splicing. Each pair is identical
through the N-terminal and catalytic domains, but encodes distinct
C-termini [175177], giving rise to proteins termed Mnk1a/b and
Mnk2a/b, where the a form is longer (Figure 6B). Mnk1a
and Mnk2a each contain a canonical MAPK-binding motif, but
this and certain other features are absent from the b forms, giving
them distinct properties. For example, whereas Mnk1a/2a are
almost exclusively cytoplasmic, Mnk1b/2b are also found in the
nucleus. Mnk2b has very low activity compared with Mnk2b
under all conditions so far tested. Mnk1a and Mnk2a also show
very distinct characteristics: Mnk1a has low basal activity, which
is markedly activated by stimulation of ERK or p38 MAPK
signalling, whereas Mnk2a has high basal activity, which is hardly
increased by activation of these pathways and is rather insensitive
to their blockade [178,179]. One probable reason for this is that
Mnk2a, but not Mnk1a, stably binds the active phosphorylated
form of ERK [178] which can thus continuously activate Mnk2a,
keeping it in an active form. In fact, when complexed with
Mnk2a, phospho-ERK is protected from dephosphorylation
by MAPK phosphatases [178]; this may explain why Mnk2a
activity is high even in cells where ERK has not been acutely
activated or which have been treated with inhibitors of the ERK
pathway. In cells which express Mnk2a, the activation of mTOR
signalling may promote eIF4E phosphorylation by increasing the
association of eIF4E with eIF4G/Mnk2, even in response to agents
that do not turn on ERK or p38 MAPK /.
The physiological significance of eIF4E phosphorylation in
mammals remains unclear, although several possibilities have
been discussed [180]. In Drosophila, mutation of the residue corresponding to Ser209 (Ser251 in deIF4E) leads to decreased viability
and impaired growth and development [181]. For example, by
weakening the interaction of eIF4E with the cap, phosphorylation
of eIF4E may allow the eIF4F complex (and associated factors,
such as eIF3 and the 40 S subunit) to detach from the 5 -cap during
scanning, perhaps accelerating this process or allowing a second
initiation complex to bind to the mRNA even before the previous
one has located the start codon. This could be especially important
for the efficient translation of mRNAs with long 5 -UTRs.
Alternatively, phosphorylation of eIF4E may allow it to disengage from mRNAs that are already being translated in order to
bind to others that have arrived recently in the cytoplasm or have
perhaps been translationally derepressed to allow their translation.
It is notable that eIF4E phosphorylation is generally mediated by

Figure 7

Control of ARE-containing mRNAs

The coding region (flanked by AUG and stop codons), the 5 -cap (star) and the poly(A) tail are
indicated, as are the AREs in the 3 -UTR. ARE-BPs bind to the AREs, inducing destabilization and
probably also impairing translation of the mRNA. Certain ARE-BPs are substrates for kinases in
downstream of the ERK and especially the p38 MAPK pathways. Phosphorylation of the ARE-BPs
may alter their affinity for the mRNA or otherwise remodel the mRNAprotein complexes,
facilitating the stabilization of the mRNAs or their translation, thereby promoting production of
the encoded protein. Probably the most widely studied such mRNA is that for TNF.

mitogen- or stress- and cytokine-activated signalling rather than


by anabolic signals, such as insulin. The former type of signals
may elicit the activation of the translation of new subsets of
mRNAs, and, as suggested, eIF4E phosphorylation may contribute to this.
Data from Aplysia californica indicate that the dephosphorylation of eIF4E triggers a switch from cap-dependent to cap-independent translation, which involves an IRES (internal ribosomeentry site) [182]. IRES elements are also found in mRNAs from
many viruses that infect mammalian cells, but no role for eIF4E
(de)phosphorylation in modulating their translation has yet been
reported. However, Mnks have been linked to modulation of the
translation of certain viral RNAs, such as HSV-1 (herpes simplex
virus-1) (where the use of the Mnk inhibitor CGP57380 revealed
a positive role for the Mnks in HSV-1 mRNA translation and viral
replication [183]) and adenovirus (where an adenovirus-encoded
protein of 100 kDa displaces Mnks from eIF4G, thus impairing
eIF4E phosphorylation). This is associated with impairment
of host cell translation without inhibiting adenoviral mRNA
translation, which is initiated via a cap-independent mechanism
(but not via an IRES [184]).
In addition to eIF4E, Mnks also phosphorylate eIF4G ([171],
and J. L. Parra-Palau, M. Buxade and C. G. Proud, unpublished
work) (Figure 6A) and a few other proteins, including at least one
that binds to the regulatory region of cytokine mRNAs [hnRNP
(heterogeneous nuclear ribonucleoprotein) A1] [169].
Regulation of ARE-containing mRNAs by MAPK signalling

Recent data indicate that the Mnks play a key role in the production of the inflammatory cytokine, TNF [168,169] (Figures 6A
and 7). Overexpression of TNF leads to inflammatory diseases

c The Authors Journal compilation 
c 2007 Biochemical Society

228

C. G. Proud

well-established that one of these, TTP (tristetraprolin), is


phosphorylated by MK2 [191]. Recent data indicate that, via
TTP, MK2 regulates both the stability and the translation of
the TNF mRNA [192]. MK2 also phosphorylates hnRNP A0
(another ARE-BP) [166] and PABP-1 [193].
The available data suggest that several ARE-BPs may be Mnk
substrates, including hnRNP A1, which has been shown to be
a physiological Mnk substrate [169,194]. Phosphorylation of
hnRNP A1 by Mnks decreases its ability to bind the TNF
mRNA AREs in vitro and the TNF mRNA in cells [169]. This
suggests that hnRNP A1 may impair the expression of TNF by
inhibiting the translation of the TNF mRNA and/or by mediating
its sequestration into stress granules, sites for storage/triage of
mRNAs that are not actively being translated [195,196].
Although use of CGP57380 fails to distinguish between Mnk1
and Mnk2, the observations that Mnk1 activity is tightly regulated,
whereas that of Mnk2 is not [179], and that siRNA-mediated
knockdown of Mnk1 alone markedly impairs TNF synthesis
[169], suggest that Mnk1 is likely to be the more important regulator here. The Mnks and MK2 may be promising new targets for
the development of novel anti-inflammatory drugs. In view of the
highly unusual nature of the ATP-binding pocket in the Mnks,
there may be scope for developing ATP-competitive agents that
are Mnk-specific [197].
The RSKs (p90RSK s) and translational control
Figure 8 Detailed overview of the control of protein synthesis by signal
transduction pathways
This diagram provides a fuller overview of the links between signalling pathways and the control
of the translational machinery, extending the simplified scheme shown in Figure 1. For details,
see the text. Steps which are unclear or poorly defined are indicated by question marks. Key
regulators of the translational machinery or of the translation of specific mRNAs are boxed.
RNA-BP, RNA-binding protein.

[185], and mice expressing a TNF mRNA that lacks the AREs
develop chronic arthritis and bowel inflammation similar to
Crohns disease [185]. Accordingly, TNF production is under
very tight control which is exerted at multiple levels: transcription,
splicing, mRNA stability and mRNA translation [186,187]. Posttranscriptional control of TNF synthesis is largely mediated
through AREs in the 3 -UTR of its mRNA [188]. Indeed, mRNAs
for many cytokines, proteins that modulate cell proliferation or
growth, and products of immediate early genes contain AREs that
modulate mRNA stability or translation.
Signalling through the ERK and p38 MAPK / pathways
promotes TNF expression, suggesting a role for these enzymes,
or for kinases that are activated by them, in this regulation.
Indeed, data from overexpression studies, use of siRNA (small
interfering RNA) and of a selective Mnk inhibitor point to a key
positive role for Mnks in the production of TNF in Jurkat T-cells
[169]. For example, the Mnk inhibitor CGP57380 or siRNAmediated knockdown of Mnk1 greatly inhibits TNF synthesis
[168,169]. These inhibitory effects do not seem to be exerted
through decreases in the level of the TNF mRNA, suggesting
they may reflect impairment of the translation of the mRNA.
It is now well established that MK2, an enzyme that is activated
by p38 MAPK /, plays a key role in TNF production at a posttranscriptional level [189] (Figure 8). Mice lacking MK2 show,
for example, a reduced response to endotoxin [189] and resistance
to collagen-induced arthritis, a process in which TNF plays a
critical role [190]. The AREs of the 3 -UTRs of the mRNAs
for cytokines such as TNF bind a number of specific proteins
[called ARE-BPs (ARE-binding proteins)] [188]. It is now

c The Authors Journal compilation 
c 2007 Biochemical Society

The protein kinases termed RSKs (or p90RSK s) are also activated
by ERK signalling (but not by p38 MAPKs) [198]. They may provide a link between Ras/Raf/MEK (MAPK/ERK kinase)/ERK
signalling and the activation of mTOR, thus providing further
examples of the mechanisms by which oncogenes may control
mTOR. mTORC1 signalling is activated by signalling through
MEK/ERK. RSKs can phosphorylate TSC2 at a C-terminal site,
Ser1798 in the most widely used numbering system [199,200] (Figures 5 and 6). As for the phosphorylation of TSC2 by PKB, there
is a paucity of direct evidence that RSK-mediated phosphorylation actually impairs TSC2s Rheb-GAP. However, the circumstantial data can be interpreted as indicating that RSK-mediated
phosphorylation of TSC2 leads to activation of mTOR. This
would allow growth factors and mitogens, and, in cardiomyocytes,
hypertrophic 1 -adrenergic agonists, to turn on mTORC1 signalling (to the S6Ks, 4E-BPs and eEF2 kinase). This link could of
course be mediated by other kinases in the pathway, including
ERKs themselves, as suggested by Ma et al. [201]. These authors
identified Ser664 as the major site in TSC2 that is phosphorylated by ERK in vitro, and showed that mutation of this site (together with another, Ser540 ) to alanine increased the ability of
TSC2 to suppress mTORC1 signalling in MEFs, Ras-induced
transformation and tumorigenesis in a model of TSC-linked tumours. These and other data suggest that phosphorylation of TSC2
by ERK links Ras/Raf/MEK signalling to the control of mTORC1.
However, Rolfe et al. [199] were unable to observe any phosphorylation of TSC2 by ERK2 in vitro. Furthermore, in their
quantitative (mass spectrometric) phosphorylation study, Ballif
et al. [202] only observed an increase in phosphorylation of one
site in TSC2 that matches the minimal consensus for phosphorylation by ERK (Ser/Thr-Pro; Ser664 ), and the changes here were
both modest in magnitude and slow in onset. It thus remains to
be firmly established whether activation of mTORC1 signalling
via the ERK pathway is mediated through the phosphorylation of
TSC2 directly by ERK or by a downstream kinase such as RSK.
p90RSK also phosphorylates at least two other proteins that are
involved in translational control. It phosphorylates eEF2 kinase at
Ser366 , the same site as S6K1, which inhibits eEF2 kinase activity

Signal transduction pathways and the control of protein synthesis

[50]. This may underlie, at least in part, the ability of agents that
activate ERK signalling to induce the dephosphorylation of eEF2
[203]. Similarly, p90RSK and S6K1 phosphorylate eIF4B at the
same site, Ser422 , promoting its association with eIF3 [51].
Bearing in mind that ERK/p90RSK signalling also leads to activation of mTORC1, PI3K/PKB and Ras/ERK signalling clearly
converge on a number of components of the translational machinery to promote the assembly of initiation factor complexes and
the activation of the elongation machinery. This is likely to be a
key element of the abilities of agents that stimulate these pathways to promote cell growth and proliferation. Both pathways lie
downstream of proteins that are implicated in tumorigenesis or
tumour suppression, e.g. PI3K and PTEN, in one case, and Ras
and Raf, in the other.
Other stress conditions

Other stressful conditions impair translation and affect multiple


components of the translational machinery [204]. The conditions
that are often used as cell stresses, such as treatment with arsenite
or H2 O2 (oxidative stress), heat shock or hyperosmolarity probably exert multiple effects. These can include the activation of
stress-activated MAPKs, especially p38 MAPK. Stresses can also
elicit the stimulation of certain eIF2 kinases, e.g. PERK [PKR
(protein kinase R)-like endoplasmic reticulum kinase] and HRI
(haem-regulated inhibitor), but that topic lies beyond the scope of
the present review.
As described above, p38 MAPKs / activate the Mnks
(mainly Mnk1a, [176,178,179]). p38 MAPKs are also activated
by endotoxin, cytokines and other immune modulators. However,
the significance of this for the control of translation is unclear:
they may reflect roles in the control of protein synthesis by such
stimuli, but this requires detailed study.
CONCLUSIONS AND PERSPECTIVES

This review has described a range of regulatory inputs that impinge on the basal translational machinery, or on the translation
of specific messages, as summarized schematically in Figure 8.
Indeed, the last few years have seen major advances in our understanding of the control of the translational apparatus and of the
signalling pathways, such as mTOR, that modulate it.
It is clear that the initiation and elongation stages of translation
are controlled by many regulatory inputs, but their quantitative
importance for the overall regulation of protein synthesis remains
to be established. This may vary between cell types and between
stimuli, and be overlaid with the effects of nutrients and cellular
energy status. More complete information on the roles of individual components come from studies on mice in which individual
factors have been knocked out (for components that are not essential, e.g. the 4E-BPs) or mutated (knock-ins, for key proteins such
as S6 [126] and eIF2 [205]). A particularly exciting aspect is the
realization that dysfunction or dysregulation of the translational
machinery leads to human diseases as diverse as cancer, tissue
hypertrophy, neurodegeneration and inflammation. One hopes
that further discoveries related to the mechanisms and control of
mRNA translation will help our understanding of these conditions
and ultimately open up new therapeutic approaches for them.
Note added in proof (received 26 February 2007)

While this paper was under review, Le Bacquer et al. [216]


reported that mice lacking 4E-BP1 and 4E-BP2 show a phenotype
involving diet-induced obesity and insulin resistance.

229

Recent work in the my laboratory on the control of mRNA translation has been supported
by the Wellcome Trust, the U.K. Medical Research Council (MRC), the U.K. Biotechnology
and Biological Sciences Research Council (BBSRC), the British Heart Foundation (BHF),
the European Union (EU), the Signal Transduction Therapy Consortium at the University
of Dundee, the Heart and Stroke Foundation of Canada, the Canadian Institutes for Health
Research, and Ajinomoto Amino Acid Research Program. I apologize to the authors of
those research papers whose original contributions I was unable to cite, owing to space
considerations. In many cases, recent review articles have been cited instead.

REFERENCES
1 Hannan, R. D., Jenkins, A., Jenkins, A. K. and Brandenburger, Y. (2003) Cardiac
hypertrophy: a matter of translation. Clin. Exp. Pharmacol. Physiol. 30, 517527
2 Inoki, K., Corradetti, M. N. and Guan, K. L. (2005) Dysregulation of the TSCmTOR
pathway in human disease. Nat. Genet. 37, 1924
3 Conlon, I. and Raff, M. (2003) Differences in the way a mammalian cell and yeast cells
coordinate cell growth and cell-cycle progression. J. Biol. 2, 7
4 Hinnebusch, A. G. (2000) Mechanism and regulation of methionyl-tRNA binding to
ribosomes. In Translational Control of Gene Expression (Sonenberg, N., Hershey,
J. W. B. and Mathews, M. B., eds.), pp. 185243, Cold Spring Harbor Laboratory Press,
Cold Spring Harbor
5 Pestova, T. V., Lorsch, J. R. and Hellen, C. U. T. (2006) The mechanism of translation
initiation in eukaryotes. In Translational Control in Biology and Medicine (Mathews,
M. B., Sonenberg, N. and Hershey, J. W. B., eds.), pp. 87128, Cold Spring Harbor
Laboratory Press
6 Gomez, E., Mohammad, S. S. and Pavitt, G. D. (2002) Characterization of the minimal
catalytic domain within eIF2B: the guanine-nucleotide exchange factor for translation
initiation. EMBO J. 21, 52925301
7 Wang, X., Paulin, F. E. M., Campbell, L. E., Gomez, E., OBrien, K., Morrice, N. and
Proud, C. G. (2001) Eukaryotic initiation factor 2B: identification of multiple
phosphorylation sites in the subunit and their roles in vivo . EMBO J. 20, 43494359
8 Welsh, G. I. and Proud, C. G. (1993) Glycogen synthase kinase-3 is rapidly inactivated
in response to insulin and phosphorylates eukaryotic initiation factor eIF-2B.
Biochem. J. 294, 625629
9 Dever, T. E., Dar, A. C. and Sicheri, F. (2006) The eIF2 kinases. In Translational Control
in Biology and Medicine (Mathews, M. B., Sonenberg, N. and Hershey, J. W. B., eds.),
pp. 319344, Cold Spring Harbor Laboratory Press, Cold Spring Harbor
10 Ron, D. and Harding, H. P. (2006) eIF2 phosphorylation in cellular stress responses
and disease. In Translational Control in Biology and Medicine (Mathews, M. B.,
Sonenberg, N. and Hershey, J. W. B., eds.), pp. 345368, Cold Spring Harbor
Laboratory Press, Cold Spring Harbor
11 Hardt, S. E., Tomita, H., Katus, H. A. and Sadoshima, J. (2004) Phosphorylation of
eukaryotic translation initiation factor 2B by glycogen synthase kinase-3 regulates
-adrenergic cardiac myocyte hypertrophy. Circ. Res. 94, 926935
12 Kleijn, M., Welsh, G. I., Scheper, G. C., Voorma, H. O., Proud, C. G. and Thomas,
A. A. M. (1998) Nerve and epidermal growth factors induce protein synthesis and eIF2B
activation in PC12 cells. J. Biol. Chem. 273, 55365541
13 Welsh, G. I., Miyamoto, S., Proud, C. G. and Safer, B. (1996) T-cell activation leads to
rapid stimulation of translation initiation factor eIF-2B and inactivation of glycogen
synthase kinase-3. J. Biol. Chem. 271, 1141011413
14 Welsh, G. I. and Proud, C. G. (1992) Regulation of protein synthesis in Swiss 3T3
fibroblasts: rapid activation of the guanine-nucleotide-exchange factor by insulin and
growth factors. Biochem. J. 284, 1923
15 Welsh, G. I., Miller, C. M., Loughlin, A. J., Price, N. T. and Proud, C. G. (1998)
Regulation of eukaryotic initiation factor eIF2B: glycogen synthase kinase-3
phosphorylates a conserved serine which undergoes dephosphorylation in response to
insulin. FEBS Lett. 421, 125130
16 Leegwater, P. A. J., Vermeulen, G., Koenst, A. A. M., Naidu, S., Mulders, J., Visser, A.,
Kersbergen, P., Mobach, D., Fonds, D. and van Berkel, C. G. M. (2001) Subunits of the
translation initiation factor eIF2B are mutated in leukoencephaly with vanishing white
matter. Nat. Genet. 29, 383388
17 Scheper, G. C., Proud, C. G. and Van der Knaap, M. S. (2006) Defective translation
initiation causes vanishing of cerebral white matter. Trends Mol. Med. 12, 159166
18 Fogli, A. and Boespflug-Tanguy, O. (2006) The large spectrum of eIF2B-related
diseases. Biochem. Soc. Trans. 34, 2229
19 Li, W., Wang, X., Van der Knaap, M. S. and Proud, C. G. (2004) Mutations linked to
leukoencephalopathy with vanishing white matter impair the function of the eukaryotic
initiation factor 2B complex in diverse ways. Mol. Cell. Biol. 24, 32953306
20 Richardson, J. P., Mohammad, S. S. and Pavitt, G. D. (2004) Mutations causing
childhood ataxia with central nervous system hypomyelination reduce eukaryotic
initiation factor 2B complex formation and activity. Mol. Cell. Biol. 24, 23522363

c The Authors Journal compilation 
c 2007 Biochemical Society

230

C. G. Proud

21 Minich, W. B., Balasta, M. L., Goss, D. J. and Rhoads, R. E. (1994) Chromatographic


resolution of in vivo phosphorylated and nonphosphorylated eukaryotic translation
initiation factor eIF-4E: increased cap affinity of the phosphorylated form.
Proc. Natl. Acad. Sci. U.S.A. 91, 76687672
22 Scheper, G. C., van Kollenburg, B., Hu, J., Luo, Y., Goss, D. J. and Proud, C. G. (2002)
Phosphorylation of eukaryotic initiation factor 4E markedly reduces its affinity for
capped mRNA. J. Biol. Chem. 277, 33033309
23 Slepenkov, S. V., Darzynkiewicz, E. and Rhoads, R. E. (2006) Stopped-flow kinetic
analysis of eIF4E and phosphorylated eIF4E binding to cap analogs and capped
oligoribonucleotides: evidence for a one-step binding mechanism. J. Biol. Chem. 281,
1492714938
24 Gingras, A.-C., Raught, B. and Sonenberg, N. (1999) eIF4 translation factors: effectors of
mRNA recruitment to ribosomes and regulators of translation. Annu. Rev. Biochem. 68,
913963
25 Imataka, H., Gradi, A. and Sonenberg, N. (1998) A newly identified N-terminal amino
acid sequence of human eIF4G binds poly(A)-binding protein and functions in
cap-dependent initiation. EMBO J. 17, 74807489
26 Harris, T. E., Chi, A., Shabanowitz, J., Hunt, D. F., Rhoads, R. E. and Lawrence, Jr, J. C.
(2006) mTOR-dependent stimulation of the association of eIF4G and eIF3 by insulin.
EMBO J. 25, 16591668
27 Mader, S., Lee, H., Pause, A. and Sonenberg, N. (1995) The translation initiation factor
eIF-4E binds to a common motif shared by the translation factor eIF-4 and the
translational repressors 4E-binding proteins. Mol. Cell. Biol. 15, 49904997
28 Le Sourd, S. F., Boulben, S., Le, B. R., Cormier, P., Morales, J., Belle, R. and
Mulner-Lorillon, O. (2006) eEF1B: at the dawn of the 21st century.
Biochim. Biophys. Acta 1759, 1331
29 Jorgensen, R., Merrill, A. R. and Andersen, G. R. (2006) The life and death of translation
elongation factor 2. Biochem. Soc. Trans. 34, 16
30 Price, N. T., Redpath, N. T., Severinov, K. V., Campbell, D. G., Russell, J. M. and Proud,
C. G. (1991) Identification of the phosphorylation sites in elongation factor-2 from rabbit
reticulocytes. FEBS Lett. 282, 253258
31 Carlberg, U., Nilsson, A. and Nygard, O. (1990) Functional properties of phosphorylated
elongation factor 2. Eur. J. Biochem. 191, 639645
32 Ryazanov, A. G. (2002) Elongation factor-2 kinase and its newly discovered relatives.
FEBS Lett. 514, 2629
33 Ryazanov, A. G., Pavur, K. S. and Dorovkov, M. V. (1999) Alpha kinases: a new class of
protein kinases with a novel catalytic domain. Curr. Biol. 9, R43R45
34 Yamaguchi, H., Matsushita, M., Nairn, A. C. and Kuriyan, J. (2001) Crystal structure of
the atypical protein kinase domain of a TRP channel with phosphotransferase activity.
Mol. Cell 7, 10471057
35 Herbert, T. P. and Proud, C. G. (2006) Regulation of translation elongation and the
cotranslational protein targeting pathway. In Translational Control in Biology and
Medicine (Mathews, M. B., Sonenberg, N. and Hershey, J. W. B., eds.), pp. 601624,
Cold Spring Harbor Laboratory Press, Cold Spring Harbor
36 Diggle, T. A., Subkhankulova, T., Lilley, K. S., Shikotra, N., Willis, A. E. and Redpath,
N. T. (2001) Phosphorylation of elongation factor-2 kinase on serine 499 by
cAMP-dependent protein kinase induces Ca2+ /calmodulin-independent activity.
Biochem. J. 353, 621626
37 Redpath, N. T. and Proud, C. G. (1993) Cyclic AMP-dependent protein kinase
phosphorylates rabbit reticulocyte elongation factor-2 kinase and induces
calcium-independent activity. Biochem. J. 293, 3134
38 Diggle, T. A., Redpath, N. T., Heesom, K. J. and Denton, R. M. (1998) Regulation of
protein synthesis elongation factor-2 kinase by cAMP in adipocytes. Biochem. J. 336,
525529
39 Hovland, R., Eikhom, T. S., Proud, C. G., Cressey, L. I., Lanotte, M., Doskeland, S. O. and
Houge, G. (1999) cAMP inhibits translation by inducing Ca2+ /calmodulin-independent
elongation factor 2 kinase activity in IPC-81 cells. FEBS Lett. 444, 97101
40 Scheetz, A. J., Nairn, A. C. and Constantine-Paton, M. (2000) NMDA-mediated control
of protein synthesis at developing synapses. Nat. Neurosci. 3, 17
41 Scheetz, A. J., Nairn, A. C. and Constantine-Paton, M. (1997) N -methyl-D-aspartate
receptor activation and visual activity induce elongation factor-2 phosphorylation in
amphibian tecta: a role for N -methyl-D-aspartate receptors in controlling protein
synthesis. Proc. Natl. Acad. Sci. U.S.A. 94, 1477014775
42 Walden, W. E. and Thach, R. E. (1986) Translational control of gene expression in a
normal fibroblast: characterization of a subclass of mRNAs with unusual kinetic
properties. Biochemistry 25, 20332041
43 Carroll, M., Warren, O., Fan, X. and Sossin, W. S. (2004) 5-HT stimulates eEF2
dephosphorylation in a rapamycin-sensitive manner in Aplysia neurites. J. Neurochem.
90, 14641476
44 Inamura, N., Nawa, H. and Takei, N. (2005) Enhancement of translation elongation in
neurons by brain-derived neurotrophic factor: implications for mammalian target of
rapamycin signaling. J. Neurochem. 95, 14381445

c The Authors Journal compilation 
c 2007 Biochemical Society

45 Klann, E. and Richter, J. D. (2006) Protein synthesis and translational control in synaptic
plasticity and learning and memory. In Translational Control in Biology and Medicine
(Mathews, M. B., Sonenberg, N. and Hershey, J. W. B., eds.), pp. 485506,
Cold Spring Harbor Laboratory Press, Cold Spring Harbor
46 Maurin, A. C., Jousse, C., Averous, J., Parry, L., Bruhat, A., Cherasse, Y., Zeng, H.,
Zhang, Y., Harding, H. P., Ron, D. and Fafournoux, P. (2005) The GCN2 kinase biases
feeding behavior to maintain amino acid homeostasis in omnivores. Cell Metab. 1,
273277
47 Avruch, J., Belham, C., Weng, Q., Hara, K. and Yonezawa, K. (2001) The p70 S6
kinase integrates nutrient and growth signals to control translational capacity.
Prog. Mol. Subcell. Biol. 26, 115154
48 Holz, M. K., Ballif, B. A., Gygi, S. P. and Blenis, J. (2005) mTOR and S6K1 mediate
assembly of the translation preinitiation complex through dynamic protein interchange
and ordered phosphorylation events. Cell 123, 569580
49 Raught, B., Peiretti, F., Gingras, A. C., Livingstone, M., Shahbazian, D., Mayeur, G. L.,
Polakiewicz, R. D., Sonenberg, N. and Hershey, J. W. (2004) Phosphorylation of
eucaryotic translation initiation factor 4B Ser422 is modulated by S6 kinases. EMBO J.
23, 17611769
50 Wang, X., Li, W., Williams, M., Terada, N., Alessi, D. R. and Proud, C. G. (2001)
Regulation of elongation factor 2 kinase by p90RSK1 and p70 S6 kinase. EMBO J. 20,
43704379
51 Shahbazian, D., Roux, P. P., Mieulet, V., Cohen, M. S., Raught, B., Taunton, J., Hershey,
J. W., Blenis, J., Pende, M. and Sonenberg, N. (2006) The mTOR/PI3K and MAPK
pathways converge on eIF4B to control its phosphorylation and activity. EMBO J. 25,
27812791
52 Wilhelm, J. E. and Smibert, C. A. (2005) Mechanisms of translational regulation in
Drosophila . Biol. Cell 97, 235252
53 Brazil, D. P., Yang, Z. Z. and Hemmings, B. A. (2004) Advances in protein kinase B
signalling: AKTion on multiple fronts. Trends Biochem. Sci. 29, 233242
54 Cully, M., You, H., Levine, A. J. and Mak, T. W. (2006) Beyond PTEN mutations: the PI3K
pathway as an integrator of multiple inputs during tumorigenesis. Nat. Rev. Cancer 6,
184192
55 Cohen, P. and Frame, S. (2001) The renaissance of GSK3. Nat. Rev. Mol. Cell Biol. 2,
769776
56 Cohen, P. and Goedert, M. (2004) GSK3 inhibitors: development and therapeutic
potential. Nat. Rev. Drug Discov. 3, 479487
57 Woods, Y., Cohen, P., Becker, W., Jakes, R., Goedert, M., Wang, X. and Proud, C. G.
(2001) DYRK phosphorylates protein synthesis initiation factor eIF2B at Ser539 and the
microtubule-associated protein Tau at Thr212 : potential role for DYRK as a GSK3 priming
kinase. Biochem. J. 355, 609615
58 Williams, D. D., Pavitt, G. D. and Proud, C. G. (2001) Characterisation of the initiation
factor eIF2B and its regulation in Drosophila melanogaster . J. Biol. Chem. 276,
37333742
59 Pap, M. and Cooper, G. M. (2001) Role of translation initiation factor 2B in the control of
cell survival by the phosphatidyl inositol 3-kinase/Akt/glycogen synthase kinase 3
signaling pathway. Mol. Cell. Biol. 22, 578586
60 Harding, H. P., Novoa, I., Zhang, Y., Zeng, H., Wek, R., Schapira, M. and Ron, D. (2000)
Regulated translation initiation controls stress-induced gene expression in mammalian
cells. Mol. Cell 6, 10991108
61 Wang, X., Janmaat, M., Beugnet, A., Paulin, F. E. M. and Proud, C. G. (2002) Evidence
that the dephosphorylation of Ser535 in the -subunit of eukaryotic initiation factor
2B is insufficient for the activation of eIF2B by insulin. Biochem. J. 367,
475481
62 Campbell, L. E., Wang, X. and Proud, C. G. (1999) Nutrients differentially modulate
multiple translation factors and their control by insulin. Biochem. J. 344, 433441
63 Kleijn, M. and Proud, C. G. (2000) The activation of eukaryotic initiation factor (eIF)2B
by growth factors in PC12 cells requires MEK/Erk signalling. FEBS Lett. 476,
262265
64 Quevedo, C., Alcazar, A. and Salinas, M. (2000) Two different signal transduction
pathways are implicated in the regulation of initiation factor 2B activity in insulin-like
growth factor-1-stimulated neuronal cells. J. Biol. Chem. 275, 1919219197
65 Quevedo, C., Salinas, M. and Alcazar, A. (2003) Initiation factor 2B activity is regulated
by protein phosphatase 1, which is activated by the mitogen-activated protein
kinase-dependent pathway in insulin-like growth factor 1-stimulated neuronal cells.
J. Biol. Chem. 278, 1657916586
66 Abraham, R. T. (2004) PI 3-kinase related kinases: big players in stress-induced
signaling pathways. DNA Repair 3, 883887
67 Wullschleger, S., Loewith, R. and Hall, M. N. (2006) TOR signaling in growth and
metabolism. Cell 124, 471484
68 Brunn, G. J., Hudson, C. C., Sekulic, A., Williams, J. M., Hosoi, H., Houghton, P. J.,
Lawrence, J. C. and Abraham, R. T. (1997) Phosphorylation of the translational repressor
PHAS-I by the mammalian target of rapamycin. Science 277, 99101

Signal transduction pathways and the control of protein synthesis


69 Burnett, P. E., Barrow, R. K., Cohen, N. A., Snyder, S. H. and Sabatini, D. M. (1998)
RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1.
Proc. Natl. Acad. Sci. U.S.A. 95, 14321437
70 Wang, X., Beugnet, A., Murakami, M., Yamanaka, S. and Proud, C. G. (2005) Distinct
signaling events downstream of mTOR cooperate to mediate the effects of amino acids
and insulin on initiation factor 4E-binding proteins. Mol. Cell. Biol. 25,
25582572
71 McMahon, L. P., Choi, K. M., Lin, T. A., Abraham, R. T. and Lawrence, Jr, J. C. (2002)
The rapamycin-binding domain governs substrate selectivity by the mammalian target of
rapamycin. Mol. Cell. Biol. 22, 74287438
72 Kimball, S. R. and Jefferson, L. S. (2006) Signaling pathways and molecular
mechanisms through which branched-chain amino acids mediate translational control of
protein synthesis. J. Nutr. 136, 227S231S
73 Sarbassov, D. D., Ali, S. M. and Sabatini, D. M. (2005) Growing roles for the mTOR
pathway. Curr. Opin. Cell Biol. 17, 596603
74 Sarbassov, D. D., Ali, S. M., Sengupta, S., Sheen, J. H., Hsu, P. P., Bagley, A. F.,
Markhard, A. L. and Sabatini, D. M. (2006) Prolonged rapamycin treatment inhibits
mTORC2 assembly and Akt/PKB. Mol. Cell 22, 159168
75 Jacinto, E., Loewith, R., Schmidt, A., Lin, S., Ruegg, M. A., Hall, A. and Hall, M. N.
(2004) Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin
insensitive. Nat. Cell Biol. 6, 11221128
76 Nojima, H., Tokunaga, C., Eguchi, S., Oshiro, N., Hidayat, S., Yoshino, K., Hara, K.,
Tanaka, J., Avruch, J. and Yonezawa, K. (2003) The mTOR partner, raptor, binds the
mTOR substrates, p70 S6 kinase and 4E-BP1, through their TOS (TOR signaling) motifs.
J. Biol. Chem. 278, 1546115464
77 Schalm, S. S., Fingar, D. C., Sabatini, D. M. and Blenis, J. (2003) TOS motif-mediated
raptor binding regulates 4E-BP1 multisite phosphorylation and function. Curr. Biol. 13,
797806
78 Hresko, R. C. and Mueckler, M. (2005) mTOR: RICTOR is the Ser473 kinase for
Akt/protein kinase B in 3T3-L1 adipocytes. J. Biol. Chem. 280, 4040640416
79 Jacinto, E., Facchinetti, V., Liu, D., Soto, N., Wei, S., Jung, S. Y., Huang, Q., Qin, J. and
Su, B. (2006) SIN1/MIP1 maintains rictormTOR complex integrity and regulates Akt
phosphorylation and substrate specificity. Cell 127, 125137
80 Sarbassov, D. D., Guertin, D. A., Ali, S. M. and Sabatini, D. M. (2005)
Phosphorylation and regulation of Akt/PKB by the rictormTOR complex. Science 307,
10981101
81 Manning, B. D. and Cantley, L. C. (2003) Rheb fills a GAP between TSC and TOR.
Trends Biochem. Sci. 28, 573576
82 Manning, B. D. and Cantley, L. C. (2003) United at last: the tuberous sclerosis complex
gene products connect the phosphoinositide 3-kinase/Akt pathway to mammalian target
of rapamycin (mTOR) signalling. Biochem. Soc. Trans. 31, 573578
83 Long, X., Lin, Y., Ortiz-Vega, S., Yonezawa, K. and Avruch, J. (2005) Rheb binds and
regulates the mTOR kinase. Curr. Biol. 15, 702713
84 Smith, E. M., Finn, S. G., Tee, A. R., Browne, G. J. and Proud, C. G. (2005) The tuberous
sclerosis protein TSC2 is not required for the regulation of the mammalian target of
rapamycin by amino acids and certain cellular stresses. J. Biol. Chem. 280,
1871718727
85 Avruch, J., Hara, K., Lin, Y., Liu, M., Long, X., Ortiz-Vega, S. and Yonezawa, K. (2006)
Insulin and amino-acid regulation of mTOR signaling and kinase activity through the
Rheb GTPase. Oncogene 25, 63616372
86 Kwiatkowski, D. J. and Manning, B. D. (2005) Tuberous sclerosis: a GAP at the
crossroads of multiple signaling pathways. Hum. Mol. Genet. 14, R251R258
87 Easton, J. B. and Houghton, P. J. (2006) mTOR and cancer therapy. Oncogene 25,
64366446
88 Jozwiak, J. (2006) Hamartin and tuberin: working together for tumour suppression.
Int. J. Cancer 118, 15
89 Altomare, D. A. and Testa, J. R. (2005) Perturbations of the AKT signaling pathway in
human cancer. Oncogene 24, 74557464
90 Guertin, D. A. and Sabatini, D. M. (2005) An expanding role for mTOR in cancer.
Trends Mol. Med. 11, 353361
91 Sabatini, D. M. (2006) mTOR and cancer: insights into a complex relationship.
Nat. Rev. Cancer 6, 729734
92 Thomas, G. V. (2006) mTOR and cancer: reason for dancing at the crossroads?
Curr. Opin. Genet. Dev. 16, 7884
93 Tee, A. R., Fingar, D. C., Manning, B. D., Kwiatkowski, D. J., Cantley, L. C. and Blenis, J.
(2002) Tuberous sclerosis complex-1 and -2 gene products function together to inhibit
mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc. Natl.
Acad. Sci. U.S.A. 99, 1357113576
94 Long, X., Ortiz-Vega, S., Lin, Y. and Avruch, J. (2005) Rheb binding to mammalian target
of rapamycin (mTOR) is regulated by amino acid sufficiency. J. Biol. Chem. 280,
2343323436

231

95 Gingras, A.-C., Gygi, S. P., Raught, B., Polakiewicz, R. D., Abraham, R. T., Hoekstra,
M. F., Aebersold, R. and Sonenberg, N. (1999) Regulation of 4E-BP1 phosphorylation:
a novel two-step mechanism. Genes Dev. 13, 14221437
96 Mothe-Satney, I., Brunn, G. J., McMahon, L. P., Capaldo, C. T., Abraham, R. T. and
Lawrence, J. C. (2000) Mammalian target of rapamycin-dependent phosphorylation of
PHAS-1 in four (S/T)P sites detected by phospho-specific antibodies. J. Biol. Chem.
275, 3383633843
97 Wang, X., Li, W., Parra, J.-L., Beugnet, A. and Proud, C. G. (2003) The C-terminus of
initiation factor 4E-binding protein 1 contains multiple regulatory features that influence
its function and phosphorylation. Mol. Cell. Biol. 23, 15461557
98 Ferguson, G., Mothe-Satney, I. and Lawrence, Jr, J. C. (2003) Ser-64 and Ser-111 in
PHAS-I are dispensable for insulin-stimulated dissociation from eIF4E. J. Biol. Chem.
278, 4745947465
99 Schalm, S. S. and Blenis, J. (2002) Identification of a conserved motif required for
mTOR signaling. Curr. Biol. 12, 632639
100 Tee, A. R. and Proud, C. G. (2002) Caspase cleavage of initiation factor 4E-binding
protein 1 yields a dominant inhibitor of cap-dependent translation and reveals a novel
regulatory motif. Mol. Cell. Biol. 22, 16741683
101 Weng, Q.-P., Kozlowski, M., Belham, C., Zhang, A., Comb, M. J. and Avruch, J. (1998)
Regulation of the p70 S6 kinase by phosphorylation in vivo : analysis using site-specific
anti-phosphopeptide antibodies. J. Biol. Chem. 273, 1662116629
102 Haghighat, A., Mader, S., Pause, A. and Sonenberg, N. (1995) Repression of
cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to
eukaryotic initiation factor-4E. EMBO J. 14, 57015709
103 Blackshear, P. J., Stumpo, D. J., Carballo, E. and Lawrence, J. C. (1997) Disruption of
the gene encoding the mitogen-regulated translational modulator PHAS-I in mice.
J. Biol. Chem. 272, 3151031514
104 Tsukiyama-Kohara, K., Poulin, F., Kohara, M., DeMaria, C. T., Cheng, A., Wu, Z.,
Gingras, A. C., Katsume, A., Elchebly, M., Spiegelman, B. M. et al. (2001) Adipose
tissue reduction in mice lacking the translational inhibitor 4E-BP1. Nat. Med. 7,
11281132
105 Banko, J. L., Poulin, F., Hou, L., DeMaria, C. T., Sonenberg, N. and Klann, E. (2005) The
translation repressor 4E-BP2 is critical for eIF4F complex formation, synaptic plasticity,
and memory in the hippocampus. J. Neurosci. 25, 95819590
106 Averous, J. and Proud, C. G. (2006) When translation meets transformation: the mTOR
story. Oncogene 25, 64236435
107 Mamane, Y., Petroulakis, E., Lebacquer, O. and Sonenberg, N. (2006) mTOR, translation
initiation and cancer. Oncogene 25, 64166422
108 Ruggero, D., Montanaro, L., Ma, L., Xu, W., Londei, P., Cordon-Cardo, C. and Pandolfi,
P. P. (2004) The translation factor eIF-4E promotes tumor formation and cooperates with
c-Myc in lymphomagenesis. Nat. Med. 10, 484
109 Wendel, H. G., De Stanchina, E., Fridman, J. S., Malina, A., Ray, S., Kogan, S.,
Cordon-Cardo, C., Pelletier, J. and Lowe, S. W. (2004) Survival signalling by Akt and
eIF4E in oncogenesis and cancer therapy. Nature 428, 332337
110 Li, S., Takasu, T., Perlman, D. M., Peterson, M. S., Burrichter, D., Avdulov, S., Bitterman,
P. B. and Polunovsky, V. A. (2003) Translation factor eIF4E rescues cells from
Myc-dependent apoptosis by inhibiting cytochrome c release. J. Biol. Chem. 278,
30153022
111 Tan, A., Bitterman, P., Sonenberg, N., Peterson, M. and Polunovsky, V. (2000) Inhibition
of Myc-dependent apoptosis by euakryotic translation initiation factor 4E requires cyclin
D1. Oncogene 19, 14371447
112 Herbert, T. P., Fahraeus, R., Prescott, A., Lane, D. P. and Proud, C. G. (2000) Rapid
induction of apoptosis mediated through peptides that bind initiation factor eIF4E.
Curr. Biol. 10, 793796
113 Rosenwald, I. B., Lazaris-Karatzas, A., Sonenberg, N. and Schmidt, E. V. (1993) Elevated
levels of cyclin D1 protein in response to increased expression of eukaryotic initiation
factor 4E. Mol. Cell. Biol. 13, 73587363
114 Shantz, L. M., Coleman, C. S. and Pegg, A. E. (1996) Expression of ornithine
decarboxylase dominant negative mutant reverses eukaryotic initiation factor-4E induced
cell transformation. Cancer Res. 56, 51365140
115 Shantz, L. M. and Pegg, A. E. (1994) Regulation of ornithine decarboxylase (ODC)
expression by translation initiation factor eIF-4E. FASEB J. 8, A1309
116 Kevil, C. G., De Benedetti, A., Payne, D. K., Coe, L. L., Laroux, F. S. and Alexander, J. S.
(1996) Translational regulation of vascular permeability factor by eukaryotic initiation
factor 4E: implications for tumor angiogenesis. Int. J. Cancer 65, 785790
117 Jiang, Y. and Muschel, R. J. (2002) Regulation of matrix metalloproteinase-9 (MMP-9)
by translational efficiency in murine prostate carcinoma cells. Cancer Res. 62,
19101914
118 Graff, J. R. and Zimmer, S. G. (2003) Translational control and metastatic progression:
enhanced activity of the mRNA cap-binding protein eIF-4E selectively enhances
translation of metastasis-related mRNAs. Clin. Exp. Metastasis 20, 265273

c The Authors Journal compilation 
c 2007 Biochemical Society

232

C. G. Proud

119 Fukuchi-Shimogori, T., Ishii, I., Kashiwagi, K., Mashiba, H., Ekimoto, J. and Igarashi, K.
(1997) Malignant transformation by overproduction of translation initiation factor eIF4G.
Cancer Res. 57, 50415044
120 Rousseau, D., Kaspar, R., Rosenwald, I., Gehrke, L. and Sonenberg, N. (1996)
Translation initiation of ornithine decarboxylase and nucleocytoplasmic transport of
cyclin D1 messenger-RNA are increased in cells overexpressing eukaryotic initiation
factor 4E. Proc. Natl. Acad. Sci. U.S.A. 93, 10651070
121 Culjkovic, B., Topisirovic, I., Skrabanek, L., Ruiz-Gutierrez, M. and Borden, K. L. (2005)
eIF4E promotes nuclear export of cyclin D1 mRNAs via an element in the 3 UTR.
J. Cell Biol. 169, 245256
122 Strudwick, S. and Borden, K. L. (2002) The emerging roles for translation factor eIF4E in
the nucleus. Differentiation 70, 1022
123 Pende, M., Um, S. H., Mieulet, V., Sticker, M., Goss, V. L., Mestan, J., Mueller, M.,
Fumagalli, S., Kozma, S. C. and Thomas, G. (2004) S6K1/ /S6K2/ mice exhibit
perinatal lethality and rapamycin-sensitive 5 -terminal oligopyrimidine mRNA
translation and reveal a mitogen-activated protein kinase-dependent S6 kinase pathway.
Mol. Cell. Biol. 24, 31123124
124 Shima, H., Pende, M., Chen, Y., Fumagalli, S., Thomas, G. and Kozma, S. C. (1998)
Disruption of the p70S6k /p85S6k gene reveals a small mouse phenotype and a new
functional S6 kinase. EMBO J. 17, 66496659
125 Montagne, J., Stewart, M. J., Stocker, H., Hafen, E., Kozma, S. C. and Thomas, G. (1999)
Drosophila S6 kinase: a regulator of cell size. Science 285, 21262129
126 Ruvinsky, I., Sharon, N., Lerer, T., Cohen, H., Stolovich-Rain, M., Nir, T., Dor, Y.,
Zisman, P. and Meyuhas, O. (2005) Ribosomal protein S6 phosphorylation is a
determinant of cell size and glucose homeostasis. Genes Dev. 19, 21992211
127 Pende, M., Kozma, S. C., Jaquet, M., Oorschot, V., Burcelin, R.,
Le Marchand-Brustel, Y., Klumperman, J., Thorens, B. and Thomas, G. (2000)
Hypoinsulinaemia, glucose intolerance and diminished -cell size in S6K1-deficient
mice. Nature 408, 994997
128 Harrington, L. S., Findlay, G. M., Gray, A., Tolkacheva, T., Wigfield, S., Rebholz, H.,
Barnett, J., Leslie, N. R., Cheng, S., Shepherd, P. R. et al. (2004) The TSC12 tumor
suppressor controls insulinPI3K signaling via regulation of IRS proteins. J. Cell Biol.
166, 213223
129 Um, S. H., Frigerio, F., Watanabe, M., Picard, F., Joaquin, M., Sticker, M., Fumagalli, S.,
Allegrini, P. R., Kozma, S. C., Auwerx, J. and Thomas, G. (2004) Absence of S6K1
protects against age- and diet-induced obesity while enhancing insulin sensitivity.
Nature 431, 200205
130 Boluyt, M. O., Zheng, J. S., Younes, A., Long, X., ONeill, L., Silverman, H., Lakatta, E. G.
and Crow, M. T. (1997) Rapamycin inhibits 1 -adrenergic receptor-stimulated cardiac
myocyte hypertrophy but not activation of hypertrophy-associated genes: evidence for
involvement of p70 S6 kinase. Circ. Res. 81, 176186
131 Shioi, T., McMullen, J. R., Tarnavski, O., Converso, K., Sherwood, M. C., Manning, W. J.
and Izumo, S. (2003) Rapamycin attenuates load-induced cardiac hypertrophy in mice.
Circulation 107, 16641670
132 McMullen, J. R., Shioi, T., Zhang, L., Tarnavski, O., Sherwood, M. C., Dorfman, A. L.,
Longnus, S., Pende, M., Martin, K. A., Blenis, J. et al. (2004) Deletion of ribosomal S6
kinases does not attenuate pathological, physiological, or insulin-like growth factor 1
receptor-phosphoinositide 3-kinase-induced cardiac hypertrophy. Mol. Cell. Biol. 24,
62316240
133 Redpath, N. T., Foulstone, E. J. and Proud, C. G. (1996) Regulation of translation
elongation factor-2 by insulin via a rapamycin-sensitive signalling pathway. EMBO J.
15, 22912297
134 Browne, G. J. and Proud, C. G. (2004) A novel mTOR-regulated phosphorylation site in
elongation factor 2 kinase modulates the activity of the kinase and its binding to
calmodulin. Mol. Cell. Biol. 24, 29862997
135 Alessi, D. R., Caudwell, F. B., Andjelkovic, M., Hemmings, B. A. and Cohen, P. (1996)
Molecular basis for the substrate specificity of protein kinase B: comparison with
MAPKAP kinase-1 and p70 S6 kinase. FEBS Lett. 399, 333338
136 Knebel, A., Morrice, N. and Cohen, P. (2001) A novel method to identify protein kinase
substrates: eEF2 kinase is phosphorylated and inhibited by SAPK4/p38. EMBO J. 20,
43604369
137 Knebel, A., Haydon, C. E., Morrice, N. and Cohen, P. (2002) Stress-induced regulation
of eEF2 kinase by SB203580-sensitive and -insensitive pathways. Biochem. J. 367,
525532
138 Meyuhas, O. and Hornstein, E. (2000) Translational control of TOP mRNAs. In
Translational Control of Gene Expression (Sonenberg, N., Hershey, J. W. B. and
Mathews, M. B., eds.), pp. 671693, Cold Spring Harbor Laboratory Press,
Cold Spring Harbor
139 Jefferies, H. B. J. and Thomas, G. (1994) Elongation factor-1 mRNA is selectively
translated following mitogenic stimulation. J. Biol. Chem. 269, 43674372

c The Authors Journal compilation 
c 2007 Biochemical Society

140 Jefferies, H. B. J., Reinhard, G., Kozma, S. C. and Thomas, G. (1994) Rapamycin
selectively represses translation of the polypyrimidine tract mRNA family. Proc. Natl.
Acad. Sci. U.S.A. 91, 44414445
141 Avni, D., Biberman, Y. and Meyuhas, O. (1997) The 5 terminal oligopyrimidine tract
confers translational control on TOP mRNAs in a cell type and sequence context
dependent manner. Nucleic Acids Res. 25, 9951001
142 Hornstein, E., Git, A., Braunstein, I., Avni, D. and Meyuhas, O. (1999) The expression of
poly(A)-binding protein gene is translationally regulated in a growth-dependent fashion
through a 5 -terminal oligopyrimidine tract motif. J. Biol. Chem. 274, 17081714
143 Tang, H., Hornstein, E., Stolovich, M., Levy, G., Livingstone, M., Templeton, D.,
Avruch, J. and Meyuhas, O. (2001) Amino acid-induced translation of TOP mRNAs is
fully dependent on phosphatidylinositol 3-kinase-mediated signalling, is partially
inhibited by rapamycin, and is independent of S6K1 and rpS6 phosphorylation.
Mol. Cell. Biol. 21, 86718683
144 Mayer, C. and Grummt, I. (2006) Ribosome biogenesis and cell growth: mTOR
coordinates transcription by all three classes of nuclear RNA polymerases. Oncogene
25, 63846391
145 Schmidt, E. V. (1999) The role of c-Myc in cellular growth control. Oncogene 18,
29882996
146 Corradetti, M. N. and Guan, K. L. (2006) Upstream of the mammalian target of
rapamycin: do all roads pass through mTOR? Oncogene 25, 63476360
147 Dennis, P. B., Jaeschke, A., Saitoh, M., Fowler, B., Kozma, S. C. and Thomas, G. (2001)
Mammalian TOR: a homeostatic ATP sensor. Science 294, 11021105
148 Hardie, D. G., Scott, J. W., Pan, D. A. and Hudson, E. R. (2003) Management of cellular
energy by the AMP-activated protein kinase system. FEBS Lett. 546, 113120
149 Inoki, K., Zhu, T. and Guan, K. L. (2003) TSC2 mediates cellular energy response to
control cell growth and survival. Cell 115, 577590
150 Brugarolas, J., Lei, K., Hurley, R. L., Manning, B. D., Reiling, J. H., Hafen, E., Witters,
L. A., Ellisen, L. W. and Kaelin, Jr, W. G. (2004) Regulation of mTOR function in
response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex.
Genes Dev. 18, 28932904
151 Corradetti, M. N., Inoki, K. and Guan, K. L. (2005) The stress-inducted proteins RTP801
and RTP801L are negative regulators of the mammalian target of rapamycin pathway.
J. Biol. Chem. 280, 97699772
152 Choo, A. Y., Roux, P. P. and Blenis, J. (2006) Mind the GAP: Wnt steps onto the
mTORC1 train. Cell 126, 834836
153 Inoki, K., Ouyang, H., Zhu, T., Lindvall, C., Wang, Y., Zhang, X., Yang, Q., Bennett, C.,
Harada, Y., Stankunas, K. et al. (2006) TSC2 integrates Wnt and energy signals via a
coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell 126,
955968
154 Browne, G. J., Finn, S. G. and Proud, C. G. (2004) Stimulation of the AMP-activated
protein kinase leads to activation of eukaryotic elongation factor 2 kinase and to its
phosphorylation at a novel site, serine 398. J. Biol. Chem. 279, 1222012231
155 Horman, S., Beauloye, C., Vertommen, D., Vanoverschelde, J. L., Hue, L. and Rider,
M. H. (2003) Myocardial ischemia and increased heart work modulate the
phosphorylation state of eukaryotic elongation factor-2. J. Biol. Chem. 278,
4197041976
156 Horman, S., Browne, G. J., Krause, U., Patel, J. V., Vertommen, D., Bertrand, L.,
Lavoinne, A., Hue, L., Proud, C. G. and Rider, M. H. (2002) Activation of AMP-activated
protein kinase leads to the phosphorylation of elongation factor 2 and an inhibition of
protein synthesis. Curr. Biol. 12, 14191423
157 McLeod, L. E. and Proud, C. G. (2002) ATP depletion increases phosphorylation of
elongation factor eEF2 in adult cardiomyocytes independently of inhibition of mTOR
signalling. FEBS Lett. 531, 448452
158 Terai, K., Hiramoto, Y., Masaki, M., Sugiyama, S., Kuroda, T., Hori, M., Kawase, I. and
Hirota, H. (2005) AMP-activated protein kinase protects cardiomyocytes against hypoxic
injury through attenuation of endoplasmic reticulum stress. Mol. Cell. Biol. 25,
95549575
159 Connolly, E., Braunstein, S., Formenti, S. and Schneider, R. J. (2006) Hypoxia inhibits
protein synthesis through a 4E-BP1 and elongation factor 2 kinase pathway controlled
by mTOR and uncoupled in breast cancer cells. Mol. Cell. Biol. 26, 39553965
160 Liu, L., Cash, T. P., Jones, R. G., Keith, B., Thompson, C. B. and Simon, M. C. (2006)
Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol. Cell
21, 521531
161 Bi, M., Naczki, C., Koritzinsky, M., Fels, D., Blais, J., Hu, N., Harding, H., Novoa, I.,
Varia, M., Raleigh, J. et al. (2005) ER stress-regulated translation increases tolerance to
extreme hypoxia and promotes tumor growth. EMBO J. 24, 34703481
162 Koritzinsky, M., Magagnin, M. G., van den Buecken, T., Seigneuric, R., Savelkouls, K.,
Dostie, J., Pyronnet, S., Kaufman, R. J., Weppler, S. A., Voncken, J. W. et al. (2006) Gene
expression during acute and prolonged hypoxia is regulated by distinct mechanisms of
translational control. EMBO J. 25, 11141125

Signal transduction pathways and the control of protein synthesis


163 Li, Y., Inoki, K., Vacratsis, P. and Guan, K. L. (2003) The p38 and MK2 kinase cascade
phosphorylates tuberin, the tuberous sclerosis 2 gene product, and enhances its
interaction with 14-3-3. J. Biol. Chem. 278, 1366313671
164 Dean, J. L., Sully, G., Clark, A. R. and Saklatvala, J. (2004) The involvement of AU-rich
element-binding proteins in p38 mitogen-activated protein kinase pathway-mediated
mRNA stabilisation. Cell. Signalling 16, 11131121
165 Ronkina, N., Kotlyarov, A., Dittrich-Breiholz, O., Kracht, M., Hitti, E., Milarski, K.,
Askew, R., Marusic, S., Lin, L. L., Gaestel, M. and Telliez, J. B. (2006) The MAPKAP
kinases MK2 and MK3 cooperate in stimulation of TNF biosynthesis and stabilization of
p38 MAPK. Mol. Cell. Biol. 27, 170181
166 Rousseau, S., Morrice, N., Peggie, M., Campbell, D. G., Gaestel, M. and Cohen, P.
(2002) Inhibition of SAPK2a/p38 prevents hnRNP A0 phosphorylation by MAPKAP-K2
and its interaction with cytokine mRNAs. EMBO J. 21, 65056514
167 Tietz, A. B., Malo, A., Diebold, J., Kotlyarov, A., Herbst, A., Kolligs, F. T.,
Brandt-Nedelev, B., Halangk, W., Gaestel, M., Goke, B. and Schafer, C. (2006) Gene
deletion of MK2 inhibits TNF- and IL-6 and protects against cerulein-induced
pancreatitis. Am. J. Physiol. Gastrointest. Liver Physiol. 290, G1298G1306
168 Andersson, K. and Sundler, R. (2006) Posttranscriptional regulation of TNF expression
via eukaryotic initiation factor 4E (eIF4E) phosphorylation in mouse macrophages.
Cytokine 33, 5257
169 Buxade, M., Parra, J. L., Rousseau, S., Shpiro, N., Marquez, R., Morrice, N., Bain, J.,
Espel, E. and Proud, C. G. (2005) The Mnks are novel components in the control of
TNF biosynthesis and phosphorylate and regulate hnRNP A1. Immunity 23, 177189
170 Fukunaga, R. and Hunter, T. (1997) Mnk1, a new MAP kinase-activated protein kinase,
isolated by a novel expression screening method for identifying protein kinase
substrates. EMBO J. 16, 19211933
171 Wang, X., Flynn, A., Waskiewicz, A. J., Webb, B. L. J., Vries, R. G., Baines, I. A.,
Cooper, J. and Proud, C. G. (1998) The phosphorylation of eukaryotic initiation factor
eIF4E in response to phorbol esters, cell stresses and cytokines is mediated by distinct
MAP kinase pathways. J. Biol. Chem. 273, 93739377
172 Waskiewicz, A. J., Flynn, A., Proud, C. G. and Cooper, J. A. (1997) Mitogen-activated
kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J. 16, 19091920
173 Pyronnet, S., Imataka, H., Gingras, A. C., Fukunaga, R., Hunter, T. and Sonenberg, N.
(1999) Human eukaryotic translation initiation factor 4G (eIF4G) recruits Mnk1 to
phosphorylate eIF4E. EMBO J. 18, 270279
174 Ueda, T., Watanabe-Fukunaga, R., Fukuyama, H., Nagata, S. and Fukunaga, R. (2004)
Mnk2 and Mnk1 are essential for constitutive and inducible phosphorylation of
eukaryotic initiation factor 4E but not for cell growth or development. Mol. Cell. Biol. 24,
65396549
175 OLoghlen, A., Gonzalez, V. M., Pineiro, D., Perez-Morgado, M. I., Salinas, M. and
Martin, M. E. (2004) Identification and molecular characterization of Mnk1b, a splice
variant of human MAP kinase-interacting kinase Mnk1. Exp. Cell Res. 299, 343355
176 Scheper, G. C., Parra, J.-L., Wilson, M. L., van Kollenburg, B., Vertegaal, A. C. O., Han,
Z.-G. and Proud, C. G. (2003) The N and C termini of the splice variants of the human
mitogen-activated protein kinase-interacting kinase Mnk2 determine activity and
localization. Mol. Cell. Biol. 23, 56925705
177 Slentz-Kesler, K., Moore, J. T., Lombard, M., Zhang, J., Hollingsworth, R. and Weiner,
M. P. (2000) Identification of the human Mnk2 gene (MKNK2) through protein
interaction with estrogen receptor . Genomics 69, 6371
178 Parra, J. L., Buxade, M. and Proud, C. G. (2005) Features of the catalytic domains and C
termini of the MAPK signal-integrating kinases Mnk1 and Mnk2 determine their differing
activities and regulatory properties. J. Biol. Chem. 280, 3762337633
179 Scheper, G. C., Morrice, N. A., Kleijn, M. and Proud, C. G. (2001) The MAP kinase
signal-integrating kinase Mnk2 is an eIF4E kinase with high basal activity in mammalian
cells. Mol. Cell. Biol. 21, 743754
180 Scheper, G. C. and Proud, C. G. (2002) Does phosphorylation of the cap-binding protein
eIF4E play a role in translation initiation? Eur. J. Biochem. 269, 53505359
181 Lachance, P. E. D., Miron, M., Raught, B., Sonenberg, N. and Lasko, P. (2002)
Phosphorylation of eukaryotic translation initiation factor 4E is critical for growth.
Mol. Cell. Biol. 22, 16561663
182 Dyer, J. R., Michel, S., Lee, W., Castellucci, V. F., Wayne, N. L. and Sossin, W. S. (2003)
An activity-dependent switch to cap-independent translation triggered by eIF4E
dephosphorylation. Nat. Neurosci. 6, 219220
183 Walsh, D. and Mohr, I. (2004) Phosphorylation of eIF4E by Mnk-1 enhances HSV-1
translation and replication in quiescent cells. Genes Dev. 18, 660672
184 Cuesta, R., Xi, Q. and Schneider, R. J. (2000) Adenovirus-specific translation by
displacement of kinase Mnk1 from cap-initiation complex eIF4F. EMBO J. 19,
34653474
185 Kontoyiannis, D., Pasparakis, M., Pizarro, T. T., Cominelli, F. and Kollias, G. (1999)
Impaired on/off regulation of TNF biosynthesis in mice lacking TNF AU-rich elements:
implications for joint and gut-associated immunopathologies. Immunity. 10, 387398

233

186 Han, J., Brown, T. and Beutler, B. (1990) Endotoxin-responsive sequences control
cachectin/tumor necrosis factor biosynthesis at the translational level. J. Exp. Med. 171,
465475
187 Neininger, A., Kontoyiannis, D., Kotlyarov, A., Winzen, R., Eckert, R., Volk, H. D.,
Holtmann, H., Kollias, G. and Gaestel, M. (2002) MK2 targets AU-rich elements and
regulates biosynthesis of tumor necrosis factor and interleukin-6 independently at
different post-transcriptional levels. J. Biol. Chem. 277, 30653068
188 Barreau, C., Paillard, L. and Osborne, H. B. (2005) AU-rich elements and associated
factors: are there unifying principles? Nucleic Acids Res. 33, 71387150
189 Kotlyarov, A., Neininger, A., Schubert, C., Eckert, R., Birchmeier, C., Volk, H. D. and
Gaestel, M. (1999) MAPKAP kinase 2 is essential for LPS-induced TNF- biosynthesis.
Nat. Cell Biol. 1, 9497
190 Hegen, M., Gaestel, M., Nickerson-Nutter, C. L., Lin, L. L. and Telliez, J. B. (2006)
MAPKAP kinase 2-deficient mice are resistant to collagen-induced arthritis. J. Immunol.
177, 19131917
191 Chrestensen, C. A., Schroeder, M. J., Shabanowitz, J., Hunt, D. F., Pelo, J. W.,
Worthington, M. T. and Sturgill, T. W. (2004) MAPKAP kinase 2 phosphorylates
tristetraprolin on in vivo sites including Ser178 , a site required for 14-3-3 binding.
J. Biol. Chem. 279, 1017610184
192 Hitti, E., Iakovleva, T., Brook, M., Deppenmeier, S., Gruber, A. D., Radzioch, D., Clark,
A. R., Blackshear, P. J., Kotlyarov, A. and Gaestel, M. (2006) Mitogen-activated protein
kinase-activated protein kinase 2 regulates tumor necrosis factor mRNA stability and
translation mainly by altering tristetraprolin expression, stability, and binding to
adenine/uridine-rich element. Mol. Cell. Biol. 26, 23992407
193 Bollig, F., Winzen, R., Gaestel, M., Kostka, S., Resch, K. and Holtmann, H. (2003) Affinity
purification of ARE-binding proteins identifies polyA-binding protein 1 as a potential
substrate in MK2-induced mRNA stabilization. Biochem. Biophys. Res. Commun. 301,
665670
194 Allemand, E., Guil, S., Myers, M., Moscat, J., Caceres, J. F. and Krainer, A. R. (2005)
Regulation of heterogeneous nuclear ribonucleoprotein A1 transport by phosphorylation
in cells stressed by osmotic shock. Proc. Natl. Acad. Sci. U.S.A. 102, 36053610
195 Anderson, P. and Kedersha, N. (2006) RNA granules. J. Cell Biol. 172, 803808
196 Guil, S., Long, J. C. and Caceres, J. F. (2006) hnRNP A1 relocalization to the stress
granules reflects a role in the stress response. Mol. Cell. Biol. 26, 57445758
197 Jauch, R., Jakel, S., Netter, C., Schreiter, K., Aicher, B., Jackle, H. and Wahl, M. C.
(2005) Crystal structures of the Mnk2 kinase domain reveal an inhibitory conformation
and a zinc binding site. Structure 13, 15591568
198 Roux, P. P. and Blenis, J. (2004) ERK and p38 MAPK-activated protein kinases: a family
of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev. 68,
320344
199 Rolfe, M., McLeod, L. E., Pratt, P. F. and Proud, C. G. (2005) Activation of protein
synthesis in cardiomyocytes by the hypertrophic agent phenylephrine requires the
activation of ERK and involves phosphorylation of tuberous sclerosis complex 2 (TSC2).
Biochem. J. 388, 973984
200 Roux, P. P., Ballif, B. A., Anjum, R., Gygi, S. P. and Blenis, J. (2004) Tumor-promoting
phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor
complex via p90 ribosomal S6 kinase. Proc. Natl. Acad. Sci. U.S.A. 101, 1348913494
201 Ma, L., Chen, Z., Erdjument-Bromage, H., Tempst, P. and Pandolfi, P. P. (2005)
Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous
sclerosis and cancer pathogenesis. Cell 121, 179193
202 Ballif, B. A., Roux, P. P., Gerber, S. A., Mackeigan, J. P., Blenis, J. and Gygi, S. P.
(2005) Quantitative phosphorylation profiling of the ERK/p90 ribosomal S6 kinasesignaling cassette and its targets, the tuberous sclerosis tumor suppressors.
Proc. Natl. Acad. Sci. U.S.A. 102, 667672
203 Wang, L. and Proud, C. G. (2002) Regulation of the phosphorylation of elongation factor
2 by MEK-dependent signalling in adult rat cardiomyocytes. FEBS Lett. 531, 285289
204 Patel, J., McLeod, L. E., Vries, R. G., Flynn, A., Wang, X. and Proud, C. G. (2002)
Cellular stresses profoundly inhibit protein synthesis and modulate the states of
phosphorylation of multiple translation factors. Eur. J. Biochem. 269, 30763085
205 Scheuner, D., Song, B., McEwen, E., Liu, C., Laybutt, R., Gillespie, P., Saunders, T.,
Bonners-Weir, S. and Kaufman, R. J. (2001) Translational control is required for the
unfolded protein response and in vivo glucose homeostasis. Mol. Cell 7, 11651176
206 Traugh, J. A. (2001) Insulin, phorbol ester and serum regulate the elongation phase of
protein synthesis. Prog. Mol. Subcell. Biol. 26, 3348
207 Herbert, T. P., Kilhams, G. R., Batty, I. H. and Proud, C. G. (2000) Distinct signalling
pathways mediate insulin and phorbol ester-stimulated eIF4F assembly and protein
synthesis in HEK 293 cells. J. Biol. Chem. 275, 1124911256
208 Colthurst, D. R., Campbell, D. G. and Proud, C. G. (1987) Structure and regulation of
eukaryotic initiation factor eIF-2: sequence of the site in the subunit phosphorylated
by the haem-controlled repressor and by the double-stranded RNA-activated inhibitor.
Eur. J. Biochem. 166, 357363

c The Authors Journal compilation 
c 2007 Biochemical Society

234

C. G. Proud

209 Joshi, B., Cai, A. L., Keiper, B. D., Minich, W. B., Mendez, R., Beach, C. M., Stolarski, R.,
Darzynkiewicz, E. and Rhoads, R. E. (1995) Phosphorylation of eukaryotic protein
synthesis initiation factor 4E at serine 209. J. Biol. Chem. 270, 1459714603
210 Raught, B., Gingras, A. C., Gygi, S. P., Imataka, H., Morino, S., Gradi, A., Aebersold, R.
and Sonenberg, N. (2000) Serum-stimulated, rapamycin-sensitive phosphorylation sites
in the eukaryotic translation initiation factor 4GI. EMBO J. 19, 434444
211 Krieg, J., Olivier, A. R. and Thomas, G. (1988) Analysis of 40 S ribosomal protein S6
phosphorylation during the mitogenic response. Methods Enzymol. 164,
575581
212 Fadden, P., Haystead, T. A. J. and Lawrence, J. C. (1997) Identification of the
phosphorylation sites in the translational regulator, PHAS-I, that are controlled by
insulin and rapamycin in rat adipocytes. J. Biol. Chem. 272, 1024010247
Received 3 January 2007/7 February 2007; accepted 8 February 2007
Published on the Internet 26 March 2007, doi:10.1042/BJ20070024


c The Authors Journal compilation 
c 2007 Biochemical Society

213 Fadden, P., Haystead, T. A. and Lawrence, J. C. (1998) Phosphorylation of the


translational regulator, PHAS-I, by protein kinase CK2. FEBS Lett. 435, 105109
214 Heesom, K., Gampel, A., Mellor, H. and Denton, R. M. (2001) Cell cycle-dependent
phosphorylation of the translational repressor eIF4E binding protein-1 (4E-BP1).
Curr. Biol. 11, 13741379
215 Merrick, W. C. and Nyborg, J. (2007) The protein biosynthesis elongation cycle. In
Translational Control of Gene Expression (Sonenberg, N., Hershey, J. W. B. and
Mathews, M. B., eds.), pp. 89125, Cold Spring Harbor Laboratory Press,
Cold Spring Harbor
216 Le Bacquer, L., Petroulakis, E., Paglialunga, S., Poulin, F., Richard, D., Cianflone, K. and
Sonenberg, N. (2007) Elevated sensitivity to diet-induced obesity and insulin resistance
in mice lacking 4E-BP1 and 4E-BP2. J. Clin. Invest. 117, 387396

You might also like