Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2021 Jan 24;288(23):6635–6651. doi: 10.1111/febs.15690

Translation initiation and its relevance in colorectal cancer

Emma Minnee 1, William James Faller 1,
PMCID: PMC9291299  PMID: 33382175

Abstract

Protein synthesis is one of the most essential processes in every kingdom of life, and its dysregulation is a known driving force in cancer development. Multiple signaling pathways converge on the translation initiation machinery, and this plays a crucial role in regulating differential gene expression. In colorectal cancer, dysregulation of initiation results in translational reprogramming, which promotes the selective translation of mRNAs required for many oncogenic processes. The majority of upstream mutations found in colorectal cancer, including alterations in the WNT, MAPK, and PI3K\AKT pathways, have been demonstrated to play a significant role in translational reprogramming. Many translation initiation factors are also known to be dysregulated, resulting in translational reprogramming during tumor initiation and/or maintenance. In this review, we outline the role of translational reprogramming that occurs during colorectal cancer development and progression and highlight some of the most critical factors affecting the etiology of this disease.

Keywords: colorectal cancer, protein synthesis, RNA translation, translation initiation, translational control


Colorectal cancer is defined by sequential mutations in a number of signaling pathways, including WNT–, MAPK–, TGFβ–, and p53–pathways. The most common mutations are in the APC and KRAS genes, causing activation of WNT– and MAPK–signaling respectively. Both of these pathways regulate mRNA translation via multiple downstream effectors, and altered protein synthesis has been shown to be vital for the development and progression of this disease.

graphic file with name FEBS-288-6635-g003.jpg


Abbreviations

4E‐BP1

eIF4E‐binding protein

5'UTR

5’‐untranslated region

APC

adenomatous polyposis coli

ATF4

activating transcription factor 4

BCL2

B‐cell lymphoma 2

CCND1

cyclin D1

CITE

cap‐independent translation enhancer

CMS

consensus molecular subtype

CRC

colorectal cancer

eIF

eukaryotic translation initiation factor

ER

endoplasmic reticulum

GCN2

general control nonderepressible 2

GEF

guanine nucleotide exchange factor

GRP78

78‐kDa glucose‐regulated protein

HRI

heme‐regulated eukaryotic initiation factor EIF‐2alpha kinase

IRES

internal ribosome entry site

ISR

integrated stress response

ITAF

IRES trans‐acting factor

KRAS

Kirsten RAS

MAPKAPK

MAPK‐activated protein kinase

MNK

MAPK‐interacting kinase

mRNA

messenger RNA

mTORC1

mechanistic target of rapamycin complex 1

ODC1

ornithine decarboxylase

PABP

poly(A)‐binding protein

PDCD4

programmed cell death 4

PERK

protein kinase R‐like endoplasmic reticulum kinase

PI3K/AKT

phosphoinositide 3‐kinase/protein kinase B

PIC

preinitiation complex

PKR

protein kinase R

RAS/MAPK

RAS/mitogen‐activated protein kinase

RP

ribosomal protein

rRNA

ribosomal RNA

RSK

p90 ribosomal S6 kinase

S6K1

70‐kDa ribosomal S6 kinase 1

SPARC

secreted protein acidic and cysteine‐rich

TC

ternary complex

uORF

upstream open reading frame

VEGFA

vascular endothelial growth factor A

WNT

Wingless and Int‐1

Introduction

Protein synthesis, via the translation of messenger RNA (mRNA), is the most energy‐consuming processes in the human body [1, 2]. It is highly regulated, and flexibility in this regulation is known to be necessary for many complex processes, such as embryonic development, cellular differentiation, and the response to stress [3, 4]. Many studies have shown that steady‐state mRNA levels are not sufficient to predict the proteome, and although there is some controversy about this, the prevailing view is that translational control provides a crucial layer of regulation in defining the ultimate cellular abundance of proteins [5, 6, 7].

The process of RNA translation can be divided into distinct phases, known as translational initiation, elongation, termination, and ribosome recycling. Initiation is considered to be the rate‐limiting phase and requires numerous individual specialized ribosomal components and eukaryotic translation initiation factors (eIFs), as described below [8].

Previous studies have revealed that the dysregulation of the translational process is a crucial driving force of tumorigenesis and progression in various types of cancer [9, 10, 11]. In order to support various oncogenic phenotypes, transformed cells alter their protein synthesis through the selective translation of specific mRNAs [11]. Indeed, almost all signaling pathways known to be important for tumorigenesis converge on translational regulation, and it may provide a promising target for therapeutic intervention [12, 13]. In colorectal cancer (CRC), alterations in WNT, c‐Myc, and KRAS signaling have all been shown to result in the dysregulation of translational, which is essential for intestinal tumorigenesis [14, 15, 16].

CRC is the third most commonly diagnosed cancer in the world. With approximately 881 000 estimated deaths each year, it is also known to be the second leading cause of cancer‐related deaths worldwide [17]. As CRC presents with a rather complex evolutionary process, combined with a heterogeneous phenotype, it is a highly difficult disease to target therapeutically [18]. In recent years, this complexity has been somewhat classified, with the stratification of CRCs into biologically defined consensus molecular subtypes (CMSs) [18, 19, 20, 21]. These studies identified four subtypes, based on a large number of clinical and biological characteristics. The relevance of this classification to dysregulated protein synthesis is largely unknown; however, two subtypes (CMS 2 and CMS 3) are primarily defined by alterations in WNT/c‐Myc signaling and KRAS mutations, respectively, both of which are directly implicated in translational control. Like most cancers, mRNA translation is commonly dysregulated in CRC [22, 23]. Indeed, the second most common alteration found in this disease is the loss of a specific rRNA modification (the m1acp3Ψ on the 18S rRNA); however, the relevance of this is currently unknown [24].

Despite decades of discovery and investigation, to date no approved treatments targeting the translation machinery are currently utilized for therapy in CRC patients [9, 25]. Since evidence points to the importance of translational control in tumor development and progression in CRC, better insight into this topic might help identify potential therapeutic targets. In this review, we will provide an overview of translational control in CRC and how this explicitly affects the etiology of this disease. Although there is still much to be understood in this subject, we will summarize and discuss the most recent findings, clarifying the overall concept of translational control in CRC. In particular, we discuss the mechanisms that regulate the translation machinery, highlighting the importance of translation initiation in translational control. Additionally, we will examine the dysregulation of translational control, focusing on the alterations found in the upstream regulating pathways, as well as in the translation machinery itself, and their role regarding tumorigenesis in CRC.

mRNA translation initiation

Together, the synthesis, accumulation, and translation of mRNA transcripts combine to regulate the cellular abundance of diverse proteins in cells, tissues, and organisms [3, 6, 26]. Translational control is vital for regulating differential gene expression, especially in response to local and systemic changes in the cellular environment, such as stress, aging, and disease [27]. In eukaryotic cells, protein synthesis can be initiated through several mechanisms, including cap‐dependent and cap‐independent translation initiation (Fig. 1) [8, 28]. The primary mode of translation initiation is cap‐dependent translation; however, this mechanism of translation is partially suppressed in response to a variety of pathophysiological stressors [29, 30]. Under such circumstances, the cell can rapidly adjust its protein synthesis, through both cap‐dependent reprogramming and the use of cap‐independent translation initiation, which promotes the upregulation of genes important for the cellular stress response [31]. It is known that the dysregulation of this process leads to alterations in the homeostasis and maintenance of the cell. Hence, the strict regulation of this cellular process is crucial [9, 10].

Fig. 1.

Fig. 1

Eukaryotic translation initiation. Schematic representation of eukaryotic translation initiation, including cap‐dependent and cap‐independent translation initiation. The primary mode of initiation is cap‐dependent translation; however, this mechanism can be suppressed in response to a variety of pathophysiological stressors. Cap‐dependent initiation involves the formation of the 43S PIC, followed by the recruitment of the mRNA template facilitated by the eIF4F complex. Once activated, the 43S PIC scans along the 5’UTR of the transcript until the initiator codon (AUG) is recognized, resulting in the release of various eIFs and the formation of the translation competent 80S ribosome. In response to specific stressors, the cell is able to adapt its gene expression pattern through reprogramming of the protein synthesis, using the cap‐independent translation initiation such as IRES‐mediated initiation. These structured mRNA sequences downstream of the 5’UTR m7G‐cap can regulate cap‐independent translation initiation via various mechanisms using both the canonical eIFs and IRES trans‐acting factors. The exact mechanisms and detailed explanations of the translation initiation machinery are provided in the text. Created with BioRender.com.

Cap‐dependent translation initiation

The predominant form of eukaryotic translation initiation requires the recognition of the m7G‐cap of the 5’‐untranslated region (5’UTR) of an mRNA transcript, followed by ribosomal scanning [8]. Various sequence elements coordinate and facilitate the guidance of at least 12 eIFs and their associated ribosomes, with the goal of forming the elongation component 80S ribosome and beginning protein synthesis [28].

Cap‐dependent translation is initiated by the formation of a multifactorial 43S preinitiation complex (PIC) via the association of the 40S ribosomal subunit with initiation factors eIF1, eIF1A, eIF3, and eIF5 and the ternary complex (TC), which itself is composed of a GTP‐bound eIF2 and Met‐tRNAi. This pre‐assembled 43S PIC is then recruited to the m7G‐cap, facilitated by eIF4F, forming the 48S preinitiation complex. eIF4F itself is a complex consisting of a mRNA 5’‐cap‐binding subunit eIF4E, the scaffold protein eIF4G, and the RNA helicase eIF4A [8, 31, 32]. As part of the 48S complex, eIF4G also facilitates the interaction of the mRNA 3’‐poly(A) tail with the poly(A)‐binding protein (PABP), which results in the circularization of the transcript in order to stabilize the mRNA and augment translation [33, 34]. Once the 48S complex binds near the preactivated m7G‐cap of the mRNA transcript, it scans along the 5’UTR toward the 3’‐end through codon–anticodon base pairing in an ATP‐dependent fashion, until the start codon (AUG) is encountered [35, 36, 37]. The recognition of the AUG codon triggers a global conformational change in the 40S ribosomal subunit, from a scanning competent to a scanning‐arrested state, which allows codon–anticodon base pairing with Met‐tRNAi. This assembly initiates the release of the phosphate from the GTP, hydrolyzed by eIF2, leading to a reduced affinity of eIF2 for the tRNA and its subsequent release [38, 39]. Immediately after the release of eIF2, eIF5B‐GTP binds to the same domain on the 48S complex, creating an increased 60S/40S‐interacting surface, which allows the recruitment of the large 60S ribosomal subunit [39, 40, 41, 42, 43]. The correct positioning of the 60S large ribosomal subunit on the initiation complex forms the elongation component 80S ribosome, which stimulates the eIF5B‐GTP hydrolysis resulting in the dissociation of eIF5B‐GTP and any other remaining initiation factors [40, 44]. This dissociation of eIF5B demarcates the end of the initiation phase of the translation, which then proceeds into translation elongation [45].

Cap‐independent translation initiation

A second form of translation initiation exists and is usually associated with the translation of mRNA transcripts following environmental and cellular stresses [46, 47]. In response to specific stressors, including nutrient deprivation, hypoxia, endoplasmic reticulum (ER) stress, and DNA damage, the cell is able to adapt its gene expression pattern through such reprogramming of protein synthesis [30]. This translational reprogramming relies on the upstream activation of assorted stress‐related kinases and phosphatases (which are specific to the stress driving the reprogramming), leading to the modulation of the phosphorylation states of key regulating eIFs, most important of which is eIF2α [48]. As a result of this phosphorylation, cap‐dependent translation is suppressed, while cap‐independent translation is largely unaffected. This results in the preferential translation of individual stress‐related mRNAs and is a mechanism the cell can use to overcome a general repression of cap‐dependent protein synthesis [8, 49]. This process is highly regulated, and its dysregulation is commonly associated with tumor formation and cancer development.

Multiple physiological stressors can induce an integrated stress response (ISR), primarily via signaling cascades that result in the activation of various eIF2α kinases, and their activation results in the phosphorylation of the eIF2 α‐subunit, eIF2α [48]. This phosphorylation increases the affinity of eIF2‐GDP for eIF2B, a guanine nucleotide exchange factor (GEF), which thereby sequesters the eIF2B GEF activity and availability required for the formation of the 43S PIC during canonical translation initiation [50, 51, 52]. This not only results in an inhibition of canonical cap‐dependent translation initiation, but also counterintuitively stimulates the cap‐dependent translation of a small subset of mRNAs encoding proteins involved in the ISR. This subset of mRNAs contain an upstream open reading frame (uORF), allowing translation initiation of mRNAs encoding stress‐related proteins, including ATF4, triggering the cell’s stress response [53].

A common alternative to cap‐dependent recruitment of the translation machinery is the use of internal ribosome entry site (IRES)‐mediated cap‐independent translation. This mechanism relies on the direct recruitment of the ribosomes to the IRES sequence [54]. IRES elements are diverse but structured mRNA sequences downstream of the 5’UTR m7G‐cap that regulate cap‐independent translation initiation via various mechanisms using both the canonical eIFs and IRES trans‐acting factors (ITAFs) [55]. The nature of IRES elements is not well understood, and it has been suggested that such structures be renamed ‘cap‐independent translation enhancers’ (CITEs) [56]. However, regardless of this debate [57, 58], evidence suggests that approximately 10% of all mRNAs contain such structures and that these elements are primarily responsible for the recruitment of the ribosomes to the internal region of the mRNA transcript [59]. In cancer, both extrinsic and intrinsic stressors, including hypoxia, nutrient deprivation, and oncogene activation, can promote the IRES‐mediated translation of a selective set of stress‐related mRNA transcripts, among which is the proto‐oncogene c‐Myc, whose activation is a common event in CRC [60].

Upstream translation initiation regulation in colorectal cancer

Tumor outgrowth depends on a selective protein synthesis that directly determines the enhanced translation of proteins required for oncogenic cellular processes [11]. Accordingly, many studies have shown that this selective protein synthesis is a consequence of the accumulation of genomic alterations in oncogenes or tumor suppressors, and CRC is no exception to this. The majority of CRC cases are initiated by the accumulation of specific mutations within the intestinal tract, eventually leading to the progression of benign polyps/adenomas into malignant carcinomas [61]. The initiating event in the majority of CRC cases is a mutation in the tumor suppressor gene APC, promoting the permanent activation of the WNT signaling pathway [62]. APC deficiency drives the transcriptional upregulation of multiple genes, including many associated with protein synthesis and the cellular stress response, accounting for a complicated multistep carcinogenic cascade to ensure tumor cell survival [63]. The dysregulated expression of these WNT target genes is thought to allow tumor outgrowth through oncogenic ‘translational reprogramming’ [64, 65, 66]. The continuous activation of WNT signaling and the reprogramming of translational control are genetic bedrocks for the majority of the CRC cases and are therefore attractive targets for further therapeutic intervention. Accordingly, a better understanding of how WNT‐related alterations might affect the translation initiation process in CRC is of great importance.

Among the most commonly described mutations found in CRC are alterations in RAS/MAPK and PI3K/AKT pathway members, driving a subsequent activation of mTOR signaling. These alterations are known to facilitate the reprogramming of the translation machinery [27, 67, 68, 69]. Besides these alterations found in the upstream signaling pathways, the altered expression and activity of numerous eIFs [70] (e.g., eIF4E, eIF4A, eIF2α), as well as the upregulation of ribosomal biogenesis [71] and factors controlling translation in response to stress [72] (e.g., GNC2 and c‐Myc), have all been shown to dysregulate the translation initiation machinery in CRC (Fig. 2). Below, we discuss the most critical mediators affecting the oncogenic translation initiation reprogramming in CRC development and progression.

Fig. 2.

Fig. 2

Translational control in CRC. Schematic representation of the translational control in CRC, including the upstream regulatory pathways and the translation initiation machinery. Factors contributing to the translational reprogramming in CRC are mutations and alterations found in the APC gene, in genes involved in RAS/MAPK signaling, including KRAS and BRAF, and in PI3K/AKT signaling, via alterations in PTEN and PI3K (marked in red). In CRC, the translational reprogramming is mainly regulated via the function and activation of the central regulators mTORC1 and c‐Myc. Besides these alterations found in the upstream signaling pathways, the altered expression and activity of numerous eIFs as well as the upregulation of components for ribosomal biogenesis and factors controlling translation in response to stress, are shown to dysregulate the translation initiation machinery in CRC. Positive regulators of the canonical translation initiation machinery are shown in green, while the negative regulators of this process are shown in blue and the individual translation initiation factors are shown in purple. Detailed explanations and descriptions about the translational control and its reprogramming in CRC are provided in the text. Created with BioRender.com.

c‐Myc as a translation initiation regulator

An early event during CRC tumorigenesis is the loss of APC, which increases global mRNA translation rates [15]. This results in the transcription of numerous WNT target genes, among which is the proto‐oncogene c‐Myc [73], which is thought to be an essential downstream regulator of WNT signaling [74]. It has been shown that c‐Myc is at least partially responsible for the translational reprogramming in CRC, leading to the oncogenic transformation [15]. Accordingly, it has been shown that ribosomal haploinsufficiency is sufficient to block oncogenic capacity of c‐Myc mouse in a model of lymphoma [75]. It is likely that this regulation of protein synthesis, as well as the c‐Myc regulation of stress responses found in CRC cells [72], ensures tumor cell survival (Fig. 3).

Fig. 3.

Fig. 3

The function of transcription factor c‐Myc in translational control. Schematic representation of the function of c‐Myc during translational reprogramming in CRC. The loss of upstream key regulator APC increases the global translation rates via the overexpression of c‐Myc, through the constitutive accumulation of β‐cat. The constitutive overexpression of c‐Myc stimulates the transcription and activation of major downstream effectors involved in the oncogenic translation initiation machinery. c‐Myc overexpression in CRC can lead to an upregulation of ribosome biogenesis and is implicated in cellular responses to stress via regulating the activity of stress‐related kinases GCN2 and PERK and that of the eIF2α/eIF2B complex, driving cap‐independent translation initiation. Detailed explanations and descriptions about the function of c‐Myc in translational control and its reprogramming in CRC are provided in the text. Created with BioRender.com.

As well as regulating translation, c‐Myc itself is known to be regulated by translation. It contains two in‐frame start codons, AUG and CUG, of which the AUG‐initiated c‐Myc isoform shows higher protein stability and activity and is therefore thought to be the more oncogenic isoform compared with its CUG‐initiated counterpart [76, 77, 78]. As a result, the constitutive overexpression of the AUG‐initiated c‐Myc eventually stimulates the transcription of major downstream effectors involved in the oncogenic translation initiation machinery [79]. This translational feedback loop results in a signaling cascade involving GCN2 and the eIF2α/eIF2B complex, driving IRES‐mediated translation initiation [72]. Aside from this alternative translation start site, c‐Myc is also known to both regulate and be regulated by, canonical, eIF4E‐mediated translation. While this has not been shown for CRC, it is known to be crucial for various other malignancies [80, 81, 82], highlighting the centrality of the interplay between c‐Myc signaling and translation in cancer development.

c‐Myc regulation of ribosome biogenesis

Ribosomes are supramolecular RNA–protein complexes, composed of roughly 80 distinct ribosomal proteins (RPs) and four different ribosomal RNAs (rRNAs), responsible for protein synthesis in every living cell [83, 84]. Compelling evidence indicates that c‐Myc modifies ribosomal biogenesis by controlling the expression of various RPs and rRNAs, thereby regulating translation and protein synthesis in general [85]. It is well known that as a consequence of c‐Myc overexpression, rRNA synthesis is hyperactivated [86]. Recent evidence has also shown that rDNA transcription and subsequent ribosomal biogenesis are elevated specifically within proliferative cells CRCs and that many RPs, including RPL15 and RPS24, are dysregulated, but these have not yet been directly linked to c‐Myc activity [87, 88, 89, 90].

c‐Myc regulation of translation initiation

In addition to ribosomal biogenesis, c‐Myc can also regulate the transcription of various translation initiation factors [25]. The upregulation of c‐Myc induces the transcription of genes encoding for proteins involved in the translation machinery, such as eIF4E, eIF4A, eIF5A, and eIF4G, via their high‐affinity c‐Myc‐binding sites [20, 91]. However, it is thought that the main role of c‐Myc in the regulation of the translation initiation machinery lies in the coordinated activity of various factors involved in IRES‐dependent translation initiation and eIF2α. In APC‐deficient CRC models, c‐Myc overexpression induces the phosphorylation of eIF2α via the eIF2α kinases GCN2 and PERK. This results in a negative feedback loop, as the expression of c‐Myc is dependent on the activity of eIF2α. The authors show that without this feedback, the cells undergo c‐Myc‐dependent apoptosis, which is prevented by the dampening of c‐Myc translation [72]. It has also been shown that the c‐Myc‐mediated phosphorylation of eIF2α, via GCN2, may also be responsible for the upregulation of the transcription factor ATF4 leading to the enhanced expression of 4E‐BP1, a negative regulator of translation [92].

mTORC1 signaling as a translation initiation regulator

mTOR is a conserved serine/threonine (Ser/Thr) protein kinase, which serves as a regulator and coordinator of proliferation and cell growth in response to numerous growth factors, environmental stimuli, cellular energy status, and nutrient/oxygen availability [93]. Over the last few decades, mapping the mTOR signaling landscape has revealed that in addition to c‐Myc, mTORC1 lies at the nexus of all the major signaling pathways driving translational control (Fig. 4) [11]. The activity of mTORC1 is known to be controlled via the upstream signaling pathways RAS/MAPK and PI3K/AKT [27]. In CRC, oncogenic signaling via alterations and mutations in those pathways promotes translational reprogramming, predominantly through the altered function of mTORC1 [22, 94, 95, 96, 97, 98]. As a result, mTORC1 is thought to be hyperactivated in most CRC cases.

Fig. 4.

Fig. 4

The upstream regulatory pathways controlling translational reprogramming in CRC. Schematic representation of the key upstream regulatory pathways regulating translational control in CRC. mTORC1 lies at the nexus of the major signaling pathways driving translational control, and its activation can be controlled via upstream signaling pathways RAS/MAPK and PI3K/AKT. In CRC, oncogenic signaling via mutations and alterations in these pathways can promote translational reprogramming, predominantly through the altered function of mTORC1 but also via the RAS/MAPK pathway directly. Factors contributing to such hyperactivated mTORC1 signaling are mutations and alterations found in the APC gene, in genes involved in RAS/MAPK signaling, including KRAS and BRAF, and in PI3K/AKT signaling, via alterations in PTEN and PI3K. Detailed explanations and abbreviations about the function of the upstream regulatory pathways in translational control and its reprogramming in CRC are provided in the text. Created with BioRender.com.

The most extensively described mTOR signaling effectors are the eIF4E‐binding proteins (4E‐BPs) [99] and 70‐kDa ribosomal S6 kinase 1 (S6K1) [100, 101]. Since the levels of phosphorylated 4E‐BPs are frequently increased in CRC, and is associated with survival outcomes, this protein is thought to be an important mediator in CRC tumorigenesis [102]. In addition to the 4E‐BPs, phosphorylation of S6K1 has also been identified as a prognostic marker for CRC and has been shown to regulate translation initiation and elongation in models of CRC [15, 103]. Furthermore, S6K1‐mediated activation of eIF4B [104] and degradation of eIF4A inhibitor PDCD4 [105] are both known to regulate translation.

MAPK signaling as a translation initiation regulator

Other Ser/Thr kinases that regulate many essential processes are the MAPKs, which signal through a module composed of conserved, sequentially acting kinases [106]. Both the RAS/ERK and the p38/MAPK pathways converge on RNA translation and are activated by a wide range of stimuli [107, 108]. The effectors of MAPK signaling, the MAPK‐activated protein kinases (MAPKAPKs), include the p90 ribosomal S6 kinases (RSKs) [109] and the MAPK‐interacting kinases (MNKs) [110]. Since the RSKs and the MNKs are directly implicated in the regulation of mRNA translation initiation, the dysregulation of the MAPK signaling via these effectors is suggested to be important during CRC tumorigenesis (Fig. 4) [111].

MNKs are MAPKAPKs that are activated in response to carcinogenic signals and cellular stress, respectively [112]. They mediate the phosphorylation of eIF4E through binding with eIF4G [113]. Unlike the overexpression of eIF4E, which directly stimulates tumor initiation, eIF4E phosphorylation facilitates tumor progression through the translation of a selective subset of mRNAs critical for extracellular remodeling, metastasis, and tumor inflammation [27, 114]. In CRC, the specific inhibition of MNK1/2, and thereby the inhibition of eIF4E phosphorylation, results in reduced tumor cell viability and tumor growth, suggesting the importance of the MNK/eIF4E axis in CRC tumorigenesis [115, 116].

Interestingly, it has recently been shown in mouse models of CRC that KRAS activation results in the rewiring of translation, via the MNK‐mediated phosphorylation of eIF4E. This results in the increased binding to, and translation of, c‐Myc, driving proliferation. The same study showed that this was a druggable weakness in these tumors, with the combination of MNK and mTOR inhibition significantly inhibiting proliferation and extending survival. That study also showed that roughly 45% of CRC patients had a high level of signaling in both of these pathways, and that this correlated with decreased survival, suggesting that this may be a viable therapeutic option [69].

Alongside the MNKs, other MAPKAPKs that are known to be important in the dysregulation of the translation initiation in CRC are the RSKs. Increasing evidence suggests that the hyperactivation of RSK1, RSK2, and RSK4 plays a significant role in the oncogenic reprogramming of the translation initiation machinery in CRC [117, 118, 119].

Dysregulation of the translation initiation machinery in colorectal cancer

Even though the upstream regulation of the translation initiation machinery has been shown to play a major role during tumorigenesis in CRC, the aberrant function of the individual eIFs is also documented to cause translational reprogramming during tumor initiation and/or maintenance. While the exact mechanism of translational reprogramming by the eIFs remains to be fully elucidated, compelling evidence suggests that the dysregulation of these components is essential to almost all oncogenic cellular processes (reviewed in Ref. [11, 70]). Moreover, since most of the eIFs exhibit increased activity in tumorigenic cells, it has been suggested that cancer cells are ‘addicted’ to the elevated translational activity [120]. For example, it has been shown that the expression of eIFs 1, 5, and 6 are correlated with CRC progression and that they may have therapeutic potential [121]. Additionally, several studies have associated expression of various eIF3 subunits with CRC survival [55, 122, 123, 124]. In this review, however, we will focus on the most commonly described alterations found in the individual components of the translation machinery and their significant role during CRC tumorigenesis. In particular, these components include the function and assembly of the eIF4F complex, the importance of key component eIF4A, and the cellular response to psychopathological stressors through eIF2α and eIF2B.

eIF4E and 4E‐BPs

Various oncogenic signaling pathways are able to enhance and alter the translation initiation machinery, predominantly through alterations in the eIF4F complex [10]. Among the most studied members of this complex are initiation factor eIF4E and its binding partner 4E‐BP, which both fulfill a crucial role during translational control in CRC. As previously described, the hyperactivation of eIF4E is by itself sufficient to drive oncogenic transformation and tumorigenesis in CRC [125]. Upstream signaling pathways can promote eIF4E hyperactivation in multiple ways [112, 114, 126, 127]. Interestingly, however, both the expression and the phosphorylation of 4E‐BP1 are increased in CRC [102, 128]. As phosphorylation is an inhibitory event, this is counterintuitive, and it is probable that the resultant modulation of translation initiation is context‐dependent.

Once activated, eIF4E overexpression enables oncogenic transformation via the translation of a select set of mRNA transcripts involved in cellular proliferation (c‐Myc [69, 129], CCND1 [130]), cell growth (ODC1 [131]) and in angiogenesis (VEGFA [132]). Alongside this, eIF4E hyperphosphorylation facilitates metastasis through epithelial‐to‐mesenchymal transition, tumor inflammation, and extracellular remodeling [133]. While hyperphosphorylation of eIF4E is shown to be a rather early event during CRC carcinogenesis [117], eIF4E overexpression is a progressive process throughout tumor development [134, 135]. Evidence suggests that the high abundance of the eIF4E protein is strongly related to the histological type of lesion, with colorectal adenocarcinomas showing the highest eIF4E expression [135]. Aside from this, the overexpression of this initiation factor is also suggested to be associated with a higher risk of metastasis [136, 137], chemoresistance [138], and poor clinical prognosis in CRC [139].

eIF4A and PDCD4

In addition to the upregulation of eIF4E, the overexpression of RNA helicase eIF4A also alters protein synthesis and this has been shown to be of great importance in cancer development [140]. Initiation factor eIF4A is considered to be the enzymatic core of the eIF4F complex and facilitates the recruitment and scanning of the 43S PIC along the mRNA 5’UTR in an ATP‐dependent manner [141, 142]. Compelling evidence suggests that the overexpression of this factor promotes the translation of many oncogenic mRNAs involved in cell proliferation, cell survival, and angiogenesis [140]. These tend to have highly structured 5’UTRs [142] and show an enrichment of G‐quadruplex structures (reviewed in Ref. [143]). The most important negative regulator of eIF4A is its binding partner PDCD4, which further limits the expression levels of eIF4A, preventing it from participating in translation initiation.

In CRC, the activity of tumor suppressor PDCD4 is found to be downregulated through its phosphorylation by S6K1, which, as a result, enhances the eIF4A availability and activity [105, 143]. This increased expression of eIF4A promotes the translation of c‐Myc, Cyclin D1, and BCL‐2 [140, 144]. Accordingly, the specific inhibition of eIF4A reduces intestinal tumor growth and CRC cell viability [145], but additionally increases various antitumorigenic effects in CRC [146, 147], underlining its importance in CRC tumorigenesis.

eIF2α and eIF2B

Another essential complex regulating translation initiation in CRC is the TC, which, in response to stressors, can promote the ISR via eIF2α phosphorylation [48]. Phosphorylation of eIF2α enhances its affinity for eIF2B, which thereby sequesters the eIF2B activity and availability required for TC formation and as a result limits translation initiation [50, 51, 52]. While it is normally only transiently activated, under oncogenic conditions its expression levels are often found to be constitutively elevated [148]. In CRC, and cancer in general, this phenomenon can be explained by an increase in various tumorigenic stimuli, such as oncogene activation, ER stress, and nutrient deprivation [149]. Counterintuitively, both the phosphorylated and nonphosphorylated forms of eIF2α are found to be upregulated in CRC; however, the functional relevance of this is not known [148, 150]. What is known is that the phosphorylation status of eIF2α determines its function global protein synthesis and the ISR, and elevated phosphorylation increases oncogenic potential in APC‐deficient models of CRC. This suggests that the key role of eIF2α is in the global inhibition of translation, but this has not been proven thus far [72, 150].

The phosphorylation status of eIF2α can be modulated via the activation of eIF2α kinases PERK, GCN2, PKR, and HRI, depending on the stimulus [48]. PKR has been shown to be increasingly expressed in CRC progression [151], but the most studied eIF2α kinases are PERK (classically activated by misfolded proteins in the ER [152]), and GCN2 (stimulated as a result of amino acid deprivation in the cell [153]). Whereas the endogenous expression of p‐eIF2α is known for its cytoprotective role and its function in cellular adaptation [149], in cancerous circumstances, the phosphorylation of eIF2α by PERK or GCN2 is thought to increase cancer cell survival, tumor growth, and angiogenesis, at least partially through the buffering of translation in order to prevent nutrient depletion [154]. In an aggressive prostate cancer mouse model, it has been shown that translation inhibition by eIF2α phosphorylation buffers the cancer cells from dramatic unrestrained increases in protein production, allowing continued tumor growth [155]. It has also been shown that such stress‐related adaptive translational responses drive plasticity in breast cancer models, through translational reprogramming and the specific regulation of stem cell‐related mRNA isoforms with alternative 5’UTRs [156].

In CRC, the transmembrane receptor PERK can phosphorylate eIF2α which can result in both the temporal inhibition of translation initiation and the translation of a small subset of mRNAs involved in response to ER stress [157], such as ATF4 [158]. The constitutive phosphorylation of eIF2α by PERK is known to result in the loss of intestinal stem cell differentiation and stemness [159], but might also play an important role in chemoresistance [160]. The BCL‐2 inhibitor SPARC has also been shown to play a role in CRC response to ER stress, through its interaction with GRP78, a master regulator of the response to ER stress. Downregulation of SPARC appears to allow CRC cells avoid apoptosis, underlining the highly regulated nature of this response [161].

GCN2 is activated in response to amino acid deprivation and is crucial for binding uncharged tRNAs [153]. The enhanced phosphorylation of eIF2α by GCN2, in combination with elevated c‐Myc and cellular stress, is an important oncogenic mechanism in APC‐deficient cells [72]. Importantly, eIF2α phosphorylation via both PERK and GCN2 enhances the expression of selected proteins, in particular that of ATF4, which enables tumor cell survival and adaptation in c‐Myc‐driven cancer types, including CRC [92, 162]. Although the function of eIF2α and eIF2B within translational control is found to be relatively clear during conditions of cell stress, their roles in tumorigenesis are still being understood. As described above, we are beginning to reveal the function of eIF2α‐mediated translational repression in cancer, but further studies are still needed.

Conclusion

Growing evidence demonstrates the importance of the regulatory mechanisms controlling RNA translation in cancer. While both initiation and elongation have been shown to be altered in cancer, its rate‐limiting phase (translation initiation) is of particular importance. Over the last few decades, many initiation factors, as well as the signaling pathways converting into the translation machinery, have been found to contribute to the development and progression of numerous human cancer types, showing the significance of strict translational control. Although the exact underlying mechanisms by which certain translation initiation factors are involved in tumorigenesis remain to be elucidated, strong progress has been made and has revealed new insights into translational reprogramming and its function as a strong oncogenic driver, especially in CRC. The elongation phase of translation has also been shown to play a vital role in intestinal cancer in mice. This is thought to be primarily involved in the initial stages of tumorigenesis, and its role in latter stages is not yet known. However, it emphasizes that the dysregulation of whole translational regulatory circuitry in CRC may serve as the foundation for pathogenesis and etiology of the disease.

Altogether, an outstanding amount of data has indicated the critical role of the translation initiation machinery and its regulatory circuitry in cancer initiation and development, with the majority showing not only its impact on cell survival, growth, and proliferation but also the cellular response to stress and angiogenesis. However, to date, no approved treatments targeting the translation machinery are utilized for therapeutic use in CRC patients, yet even though a few promising targeting strategies are advanced to clinical trials. This can partially be explained by the fact that the translation fulfills an essential and general housekeeping function in the human body, which, as consequence, could interfere with these therapeutic treatments leading to systemic toxicity. Additionally, significant knowledge, regarding the exact underlying molecular and cellular mechanisms involved in this translational reprogramming, needs to be elucidated in order to develop more successful therapeutic treatments. Accordingly, there is a need for therapeutic approaches selectively and specifically targeting proteins involved in the translational reprogramming of tumorigenic cells in the intestine. Ultimately, as research continues to obtain a more discrete understanding of the regulatory mechanisms underlying translation initiation of the cancer genome, this will open the opportunity to develop and provide novel therapeutic approaches for CRC patients.

Conflict of interest

The authors declare no conflict of interest.

Author contributions

EM and WJF conceived, wrote, and revised the manuscript.

PEER REVIEW

The peer review history for this article is available at https://publons.com/publon/10.1111/febs.15690.

[Correction added on 24 March 2021, after first online publication: URL for peer review history has been corrected.]

Acknowledgements

This work was supported by the Dutch Cancer Society (KWF), Project Number 10535.

References

  • 1. Buttgereit F & Brand MD (1995) A hierarchy of ATP‐consuming processes in mammalian cells. Biochem J 312, 163–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Liu Y, Beyer A & Aebersold R (2016) On the dependency of cellular protein levels on mRNA abundance. Cell 165, 535–550. [DOI] [PubMed] [Google Scholar]
  • 3. Kong J & Lasko P (2012) Translational control in cellular and developmental processes. Nat Rev Genet 13, 383–394. [DOI] [PubMed] [Google Scholar]
  • 4. Genuth NR & Barna M (2018) Heterogeneity and specialized functions of translation machinery: from genes to organisms. Nat Rev Genet 19, 431–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. McManus J, Cheng Z & Vogel C (2015) Next‐generation analysis of gene expression regulation – comparing the roles of synthesis and degradation. Mol Biosyst 11, 2680–2689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Schwanhäusser B, Busse D, Li N, Dittmar G, Schuchhardt J, Wolf J, Chen W & Selbach M (2011) Global quantification of mammalian gene expression control. Nature 473, 337–342. [DOI] [PubMed] [Google Scholar]
  • 7. Li JJ, Chew G‐L & Biggin MD (2017) Quantitating translational control: mRNA abundance‐dependent and independent contributions and the mRNA sequences that specify them. Nucleic Acids Res 45, gkx898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Sonenberg N & Hinnebusch AG (2009) Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136, 731–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bhat M, Robichaud N, Hulea L, Sonenberg N, Pelletier J & Topisirovic I (2015) Targeting the translation machinery in cancer. Nat Rev Drug Discov 14, 261–278. [DOI] [PubMed] [Google Scholar]
  • 10. Pelletier J, Graff J, Ruggero D & Sonenberg N (2015) Targeting the eIF4F translation initiation complex: a critical nexus for cancer development. Cancer Res 75, 250–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Truitt ML & Ruggero D (2016) New frontiers in translational control of the cancer genome. Nat Rev Cancer 16, 288–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Sendoel A, Dunn JG, Rodriguez EH, Naik S, Gomez NC, Hurwitz B, Levorse J, Dill BD, Schramek D, Molina H et al. (2017) Translation from unconventional 5′ start sites drives tumour initiation. Nature 541, 494–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ruggero D (2013) Translational control in cancer etiology. Cold Spring Harbor Perspect Biol 5, a012336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Huang D, Sun W, Zhou Y, Li P, Chen F, Chen H, Xia D, Xu E, Lai M, Wu Y et al. (2018) Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metast Rev 37, 173–187. [DOI] [PubMed] [Google Scholar]
  • 15. Faller WJ, Jackson TJ, Knight JRP, Ridgway RA, Jamieson T, Karim SA, Jones C, Radulescu S, Huels DJ, Myant KB et al. (2015) mTORC1‐mediated translational elongation limits intestinal tumour initiation and growth. Nature 517, 497–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Wiegering A, Uthe FW, Jamieson T, Ruoss Y, Hüttenrauch M, Küspert M, Pfann C, Nixon C, Herold S, Walz S et al. (2015) Targeting translation initiation bypasses signaling crosstalk mechanisms that maintain high MYC levels in colorectal cancer. Cancer Discov 5, 768–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA & Jemal A (2018) Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68, 394–424. [DOI] [PubMed] [Google Scholar]
  • 18. Linnekamp JF, van Hooff SR, Prasetyanti PR, Kandimalla R, Buikhuisen JY, Fessler E, Ramesh P, Lee KAST, Bochove GGW, de Jong JH et al. (2018) Consensus molecular subtypes of colorectal cancer are recapitulated in in vitro and in vivo models. Cell Death Differ 25, 616–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Budinska E, Popovici V, Tejpar S, D’Ario G, Lapique N, Sikora KO, Narzo AFD, Yan P, Hodgson JG, Weinrich S et al. (2013) Gene expression patterns unveil a new level of molecular heterogeneity in colorectal cancer. J Pathology 231, 63–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Muzny DM, Bainbridge MN, Chang K, Dinh HH, Drummond JA, Fowler G, Kovar CL, Lewis LR, Morgan MB, Newsham IF et al. (2012) Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P et al. (2015) The consensus molecular subtypes of colorectal cancer. Nat Med 21, 1350–1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Fujishita T, Aoki M & Taketo MM (2009) The role of mTORC1 pathway in intestinal tumorigenesis. Cell Cycle 8, 3684–3687. [DOI] [PubMed] [Google Scholar]
  • 23. Parsyan A, Hernández G & Meterissian S (2012) Translation initiation in colorectal cancer. Cancer Metast Rev 31, 387–395. [DOI] [PubMed] [Google Scholar]
  • 24. Babaian A, Rothe K, Girodat D, Minia I, Djondovic S, Milek M, Miko SES, Wieden H‐J, Landthaler M, Morin GB et al. (2020) Loss of m1acp3Ψ ribosomal RNA modification is a major feature of cancer. Cell Rep 31, 107611. [DOI] [PubMed] [Google Scholar]
  • 25. Pal I, Safari M, Jovanovic M, Bates SE & Deng C (2019) Targeting translation of mRNA as a therapeutic strategy in cancer. Curr Hematol Malig Rep 14, 219–227. [DOI] [PubMed] [Google Scholar]
  • 26. Lécuyer E, Yoshida H, Parthasarathy N, Alm C, Babak T, Cerovina T, Hughes TR, Tomancak P & Krause HM (2007) Global analysis of mRNA localization reveals a prominent role in organizing cellular architecture and function. Cell 131, 174–187. [DOI] [PubMed] [Google Scholar]
  • 27. Roux PP & Topisirovic I (2018) Signaling pathways involved in the regulation of mRNA translation. Mol Cell Biol 38, e00070‐18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Haimov O, Sinvani H & Dikstein R (2015) Cap‐dependent, scanning‐free translation initiation mechanisms. Biochim Biophys Acta 1849, 1313–1318. [DOI] [PubMed] [Google Scholar]
  • 29. Aitken CE & Lorsch JR (2012) A mechanistic overview of translation initiation in eukaryotes. Nat Struct Mol Biol 19, 568–576. [DOI] [PubMed] [Google Scholar]
  • 30. Spriggs KA, Bushell M & Willis AE (2010) Translational regulation of gene expression during conditions of cell stress. Mol Cell 40, 228–237. [DOI] [PubMed] [Google Scholar]
  • 31. Jackson RJ, Hellen CUT & Pestova TV (2010) The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol 11, 113–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Gingras A‐C, Raught B & Sonenberg N (1999) eIF4 INITIATION FACTORS: effectors of mRNA recruitment to ribosomes and regulators of translation. Annu Rev Biochem 68, 913–963. [DOI] [PubMed] [Google Scholar]
  • 33. Kahvejian A, Svitkin YV, Sukarieh R, M’Boutchou M‐N & Sonenberg N (2005) Mammalian poly(A)‐binding protein is a eukaryotic translation initiation factor, which acts via multiple mechanisms. Gene Dev 19, 104–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Gallie DR (1991) The cap and poly(A) tail function synergistically to regulate mRNA translational efficiency. Gene Dev 5, 2108–2116. [DOI] [PubMed] [Google Scholar]
  • 35. Kozak M (1984) Selection of initiation sites by eucaryotic ribosomes: effect of inserting AUG triplets upstream from the coding sequence for preproinsulin. Nucleic Acids Res 12, 3873–3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Hinnebusch AG (2011) Molecular mechanism of scanning and start codon selection in eukaryotes. Microbiol Mol Biol Rep 75, 434–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Hinnebusch AG (2017) Structural insights into the mechanism of scanning and start codon recognition in eukaryotic translation initiation. Trends Biochem Sci 42, 589–611. [DOI] [PubMed] [Google Scholar]
  • 38. Baird TD & Wek RC (2012) Eukaryotic initiation factor 2 phosphorylation and translational control in metabolism. Adv Nutrition 3, 307–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Pisareva VP & Pisarev AV (2014) eIF5 and eIF5B together stimulate 48S initiation complex formation during ribosomal scanning. Nucleic Acids Res 42, 12052–12069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Fernández IS, Bai X‐C, Hussain T, Kelley AC, Lorsch JR, Ramakrishnan V & Scheres SHW (2013) Molecular architecture of a eukaryotic translational initiation complex. Science 342, 1240585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Pestova TV, Lomakin IB, Lee JH, Choi SK, Dever TE & Hellen CUT (2000) The joining of ribosomal subunits in eukaryotes requires eIF5B. Nature 403, 332–335. [DOI] [PubMed] [Google Scholar]
  • 42. Huang BY & Fernández IS (2020) Long‐range interdomain communications in eIF5B regulate GTP hydrolysis and translation initiation. Proc National Acad Sci USA 117, 1429–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Pelletier J & Sonenberg N (2019) The organizing principles of eukaryotic ribosome recruitment. Annu Rev Biochem 88, 307–335. [DOI] [PubMed] [Google Scholar]
  • 44. Voorhees RM, Schmeing TM, Kelley AC & Ramakrishnan V (2010) The mechanism for activation of GTP hydrolysis on the ribosome. Science 330, 835–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Wang J, Johnson AG, Lapointe CP, Choi J, Prabhakar A, Chen D‐H, Petrov AN & Puglisi JD (2019) eIF5B gates the transition from translation initiation to elongation. Nature 573, 605–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Starck SR, Tsai JC, Chen K, Shodiya M, Wang L, Yahiro K, Martins‐Green M, Shastri N & Walter P (2016) Translation from the 5′ untranslated region shapes the integrated stress response. Science 351, aad3867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Shatsky IN, Dmitriev SE, Terenin IM & Andreev DE (2010) Cap‐ and IRES‐independent scanning mechanism of translation initiation as an alternative to the concept of cellular IRESs. Mol Cell 30, 285–293. [DOI] [PubMed] [Google Scholar]
  • 48. Pakos‐Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A & Gorman AM (2016) The integrated stress response. EMBO Rep 17, 1374–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Hinnebusch AG, Ivanov IP & Sonenberg N (2016) Translational control by 5′‐untranslated regions of eukaryotic mRNAs. Science 352, 1413–1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Adomavicius T, Guaita M, Zhou Y, Jennings MD, Latif Z, Roseman AM & Pavitt GD (2019) The structural basis of translational control by eIF2 phosphorylation. Nat Commun 10, 2136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Jennings MD, Kershaw CJ, Adomavicius T & Pavitt GD (2017) Fail‐safe control of translation initiation by dissociation of eIF2α phosphorylated ternary complexes. Elife 6, e24542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kenner LR, Anand AA, Nguyen HC, Myasnikov AG, Klose CJ, McGeever LA, Tsai JC, Miller‐Vedam LE, Walter P & Frost A (2019) eIF2B‐catalyzed nucleotide exchange and phosphoregulation by the integrated stress response. Science 364, 491–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Wortel IMN, van der Meer LT, Kilberg MS & van Leeuwen FN (2017) Surviving stress: modulation of ATF4‐mediated stress responses in normal and malignant cells. Trends Endocrinol Metab 28, 794–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Mitchell SA, Spriggs KA, Bushell M, Evans JR, Stoneley M, Quesne JPCL, Spriggs RV & Willis AE (2005) Identification of a motif that mediates polypyrimidine tract‐binding protein‐dependent internal ribosome entry. Gene Dev 19, 1556–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Yang Y & Wang Z (2019) IRES‐mediated cap‐independent translation, a path leading to hidden proteome. J Mol Cell Biol 11, 911–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Shatsky IN, Terenin IM, Smirnova VV & Andreev DE (2018) Cap‐independent translation: what’s in a name? Trends Biochem Sci 43, 882–895. [DOI] [PubMed] [Google Scholar]
  • 57. Kozak M (2005) A second look at cellular mRNA sequences said to function as internal ribosome entry sites. Nucleic Acids Res 33, 6593–6602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Gilbert WV (2010) Alternative ways to think about cellular internal ribosome entry. J Biol Chem 285, 29033–29038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Weingarten‐Gabbay S, Elias‐Kirma S, Nir R, Gritsenko AA, Stern‐Ginossar N, Yakhini Z, Weinberger A & Segal E (2016) Systematic discovery of cap‐independent translation sequences in human and viral genomes. Science 351, aad4939. [DOI] [PubMed] [Google Scholar]
  • 60. Cobbold LC, Spriggs KA, Haines SJ, Dobbyn HC, Hayes C, de Moor CH, Lilley KS, Bushell M & Willis AE (2008) Identification of internal ribosome entry segment (IRES)‐trans‐acting factors for the Myc family of IRESs. Mol Cell Biol 28, 40–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Fearon ER & Vogelstein B (1990) A genetic model for colorectal tumorigenesis. Cell 61, 759–767. [DOI] [PubMed] [Google Scholar]
  • 62. Schatoff EM, Leach BI & Dow LE (2017) WNT signaling and colorectal cancer. Curr Colorectal Cancer Rep 13, 101–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Coppedè F, Lopomo A, Spisni R & Migliore L (2014) Genetic and epigenetic biomarkers for diagnosis, prognosis and treatment of colorectal cancer. World J Gastroenterol 20, 943–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Clevers H & Nusse R (2012) Wnt/β‐catenin signaling and disease. Cell 149, 1192–1205. [DOI] [PubMed] [Google Scholar]
  • 65. Zhan T, Rindtorff N & Boutros M (2017) Wnt signaling in cancer. Oncogene 36, 1461–1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Anastas JN & Moon RT (2013) WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer 13, 11–26. [DOI] [PubMed] [Google Scholar]
  • 67. Liu GY & Sabatini DM (2020) mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 21, 183–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Bogaert J & Prenen H (2011) Molecular genetics of colorectal cancer. Annu Rev Pathol Mech Dis 6, 479–507. [DOI] [PubMed] [Google Scholar]
  • 69. Knight JRP, Alexandrou C, Skalka GL, Vlahov N, Pennel K, Officer L, Teodosio A, Kanellos G, Gay DM, May‐Wilson S et al. (2020) MNK inhibition sensitizes KRAS‐mutant colorectal cancer to mTORC1 inhibition by reducing eIF4E phosphorylation and c‐MYC expression. Cancer Discov CD‐20‐0652. 10.1158/2159-8290.CD-20-0652 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. de la Parra C, Walters BA, Geter P & Schneider RJ (2018) Translation initiation factors and their relevance in cancer. Curr Opin Genet Dev 48, 82–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Chester KA, Robson L, Begent RHJ, Talbot IC, Pringle JH, Primrose L, Macpherson AJS, Boxer G, Southall P & Malcolm ADB (1989) Identification of a human ribosomal protein mRNA with increased expression in colorectal tumours. Biochim Biophys Acta 1009, 297–300. [DOI] [PubMed] [Google Scholar]
  • 72. Schmidt S, Gay D, Uthe FW, Denk S, Paauwe M, Matthes N, Diefenbacher ME, Bryson S, Warrander FC, Erhard F et al. (2019) A MYC–GCN2–eIF2α negative feedback loop limits protein synthesis to prevent MYC‐dependent apoptosis in colorectal cancer. Nat Cell Biol 21, 1413–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Korinek V, Barker N, Morin PJ, van Wichen D, de Weger R, Kinzler KW, Vogelstein B & Clevers H (1997) Constitutive transcriptional activation by a β‐catenin‐Tcf complex in APC−/− colon carcinoma. Science 275, 1784–1787. [DOI] [PubMed] [Google Scholar]
  • 74. Sansom OJ, Meniel VS, Muncan V, Phesse TJ, Wilkins JA, Reed KR, Vass JK, Athineos D, Clevers H & Clarke AR (2007) Myc deletion rescues Apc deficiency in the small intestine. Nature 446, 676–679. [DOI] [PubMed] [Google Scholar]
  • 75. Barna M, Pusic A, Zollo O, Costa M, Kondrashov N, Rego E, Rao PH & Ruggero D (2008) Suppression of Myc oncogenic activity by ribosomal protein haploinsufficiency. Nature 456, 971–975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Nanbru C, Lafon I, Audigier S, Gensac M‐C, Vagner S, Huez G & Prats A‐C (1997) Alternative translation of the proto‐oncogene c‐mycby an internal ribosome entry site. J Biol Chem 272, 32061–32066. [DOI] [PubMed] [Google Scholar]
  • 77. Sato K, Masuda T, Hu Q, Tobo T, Gillaspie S, Niida A, Thornton M, Kuroda Y, Eguchi H, Nakagawa T et al. (2019) Novel oncogene 5MP1 reprograms c‐Myc translation initiation to drive malignant phenotypes in colorectal cancer. Ebiomedicine 44, 387–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Hann SR, King MW, Bentley DL, Anderson CW & Eisenman RN (1988) A non‐AUG translational initiation in c‐myc exon 1 generates an N‐terminally distinct protein whose synthesis is disrupted in Burkitt’s lymphomas. Cell 52, 185–195. [DOI] [PubMed] [Google Scholar]
  • 79. Coller HA, Grandori C, Tamayo P, Colbert T, Lander ES, Eisenman RN & Golub TR (2000) Expression analysis with oligonucleotide microarrays reveals that MYC regulates genes involved in growth, cell cycle, signaling, and adhesion. Proc Natl Acad Sci USA 97, 3260–3265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Polunovsky VA, Rosenwald IB, Tan AT, White J, Chiang L, Sonenberg N & Bitterman PB (1996) Translational control of programmed cell death: eukaryotic translation initiation factor 4E blocks apoptosis in growth‐factorrestricted fibroblasts with physiologically expressed or deregulated Myc. Mol Cell Biol 16, 6573–6581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Truitt ML, Conn CS, Shi Z, Pang X, Tokuyasu T, Coady AM, Seo Y, Barna M & Ruggero D (2015) Differential requirements for eIF4E dose in normal development and cancer. Cell 162, 59–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Tan A, Bitterman P, Sonenberg N, Peterson M & Polunovsky V (2000) Inhibition of Myc‐dependent apoptosis by eukaryotic translation initiation factor 4E requires cyclin D1. Oncogene 19, 1437–1447. [DOI] [PubMed] [Google Scholar]
  • 83. Rodnina MV & Wintermeyer W (2009) Recent mechanistic insights into eukaryotic ribosomes. Curr Opin Cell Biol 21, 435–443. [DOI] [PubMed] [Google Scholar]
  • 84. Uechi T, Tanaka T & Kenmochi N (2001) A complete map of the human ribosomal protein genes: assignment of 80 genes to the cytogenetic map and implications for human disorders. Genomics 72, 223–230. [DOI] [PubMed] [Google Scholar]
  • 85. van Riggelen J, Yetil A & Felsher DW (2010) MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer 10, 301–309. [DOI] [PubMed] [Google Scholar]
  • 86. Morcelle C, Menoyo S, Morón‐Duran FD, Tauler A, Kozma SC, Thomas G & Gentilella A (2019) Oncogenic MYC induces the impaired ribosome biogenesis checkpoint and stabilizes p53 independent of increased ribosome content. Cancer Res 79, 4348–4359. [DOI] [PubMed] [Google Scholar]
  • 87. Mao‐De L & Jing X (2007) Ribosomal proteins and colorectal cancer. Curr Genomics 8, 43–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Wang Y, Sui J, Li X, Cao F, He J, Yang B, Zhu X, Sun Y & Pu YD (2015) RPS24 knockdown inhibits colorectal cancer cell migration and proliferation in vitro. Gene 571, 286–291. [DOI] [PubMed] [Google Scholar]
  • 89. Dong Z, Jiang H, Liang S, Wang Y, Jiang W & Zhu C (2019) Ribosomal protein L15 is involved in colon carcinogenesis. Int J Med Sci 16, 1132–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Morral C, Stanisavljevic J, Hernando‐Momblona X, Mereu E, Álvarez‐Varela A, Cortina C, Stork D, Slebe F, Turon G, Whissell G et al. (2020) Zonation of ribosomal DNA transcription defines a stem cell hierarchy in colorectal cancer. Cell Stem Cell 26, 845–861.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Fernandez PC, Frank SR, Wang L, Schroeder M, Liu S, Greene J, Cocito A & Amati B (2003) Genomic targets of the human c‐Myc protein. Gene Dev 17, 1115–1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Tameire F, Verginadis II, Leli NM, Polte C, Conn CS, Ojha R, Salinas CS, Chinga F, Monroy AlexandraM FuW, Wang P et al. (2019) ATF4 couples MYC‐dependent translational activity to bioenergetic demands during tumour progression. Nat Cell Biol 21, 889–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Saxton RA & Sabatini DM (2017) mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Charitou T, Srihari S, Lynn MA, Jarboui M‐A, Fasterius E, Moldovan M, Shirasawa S, Tsunoda T, Ueffing M, Xie J et al. (2019) Transcriptional and metabolic rewiring of colorectal cancer cells expressing the oncogenic KRASG13D mutation. Br J Cancer 121, 37–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Caputo F, Santini C, Bardasi C, Cerma K, Casadei‐Gardini A, Spallanzani A, Andrikou K, Cascinu S & Gelsomino F (2019) BRAF‐mutated colorectal cancer: clinical and molecular insights. Int J Mol Sci 20, 5369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Zhou X‐P, Loukola A, Salovaara R, Nystrom‐Lahti M, Peltomäki P, de la Chapelle A, Aaltonen LA & Eng C (2002) PTEN mutational spectra, expression levels, and subcellular localization in microsatellite stable and unstable colorectal cancers. Am J Pathol 161, 439–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Roock WD, Vriendt VD, Normanno N, Ciardiello F & Tejpar S (2011) KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer. Lancet Oncol 12, 594–603. [DOI] [PubMed] [Google Scholar]
  • 98. Roy HK, Olusola BF, Clemens DL, Karolski WJ, Ratashak A, Lynch HT & Smyrk TC (2002) AKT protooncogene overexpression is an early event during sporadic colon carcinogenesis. Carcinogenesis 23, 201–205. [DOI] [PubMed] [Google Scholar]
  • 99. Hara K, Yonezawa K, Kozlowski MT, Sugimoto T, Andrabi K, Weng Q‐P, Kasuga M, Nishimoto I & Avruch J (1997) Regulation of eIF‐4E BP1 phosphorylation by mTOR. J Biol Chem 272, 26457–26463. [DOI] [PubMed] [Google Scholar]
  • 100. Ma XM & Blenis J (2009) Molecular mechanisms of mTOR‐mediated translational control. Nat Rev Mol Cell Biol 10, 307–318. [DOI] [PubMed] [Google Scholar]
  • 101. Hay N & Sonenberg N (2004) Upstream and downstream of mTOR. Gene Dev 18, 1926–1945. [DOI] [PubMed] [Google Scholar]
  • 102. Chao M‐W, Wang L‐T, Lai C‐Y, Yang X‐M, Cheng Y‐W, Lee K‐H, Pan S‐L & Teng C‐M (2015) eIF4E binding protein 1 expression is associated with clinical survival outcomes in colorectal cancer. Oncotarget 6, 24092–24104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Wiesweg M, Reis H, Köster T, Goetz M, Worm K, Herold T, Paul A, Dechêne A, Schumacher B, Markus P et al. (2018) Phosphorylation of p70 ribosomal protein S6 kinase β‐1 is an independent prognostic parameter in metastatic colorectal cancer. Clin Colorectal Cancer 17, e331–e352. [DOI] [PubMed] [Google Scholar]
  • 104. Matassa DS, Agliarulo I, Amoroso MR, Maddalena F, Sepe L, Ferrari MC, Sagar V, D’Amico S, Loreni F, Paolella G et al. (2014) TRAP1‐dependent regulation of p70S6K is involved in the attenuation of protein synthesis and cell migration: relevance in human colorectal tumors. Mol Oncol 8, 1482–1494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Long J, Yin Y, Guo H, Li S, Sun Y, Zeng C & Zhu W (2018) The mechanisms and clinical significance of PDCD4 in colorectal cancer. Gene 680, 59–64. [DOI] [PubMed] [Google Scholar]
  • 106. Widmann C, Gibson S, Jarpe MB & Johnson GL (1999) Mitogen‐activated protein kinase: conservation of a three‐kinase module from yeast to human. Physiol Rev 79, 143–180. [DOI] [PubMed] [Google Scholar]
  • 107. Pearson G, Robinson F, Gibson TB, Xu B, Karandikar M, Berman K & Cobb MH (2001) Mitogen‐activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev 22, 153–183. [DOI] [PubMed] [Google Scholar]
  • 108. Cargnello M & Roux PP (2011) Activation and function of the MAPKs and their substrates, the MAPK activated protein kinases. Microbiol Mol Biol Rep 75, 50–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Carriere A, Ray H, Blenis J & Roux PP (2008) The RSK factors of activating the Ras/MAPK signaling cascade. Front Biosci 13, 4258–4275. [DOI] [PubMed] [Google Scholar]
  • 110. Buxade M (2008) The Mnks: MAP kinase‐interacting kinases (MAP kinase signal‐integrating kinases). Front Biosci 13, 5359–5373. [DOI] [PubMed] [Google Scholar]
  • 111. Proud CG (2015) Mnks, eIF4E phosphorylation and cancer. Biochim Biophys Acta 1849, 766–773. [DOI] [PubMed] [Google Scholar]
  • 112. Waskiewicz AJ, Flynn A, Proud CG & Cooper JA (1997) Mitogen‐activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J 16, 1909–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Pyronnet S, Imataka H, Gingras A, Fukunaga R, Hunter T & Sonenberg N (1999) Human eukaryotic translation initiation factor 4G (eIF4G) recruits Mnk1 to phosphorylate eIF4E. EMBO J 18, 270–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Robichaud N, del Rincon SV, Huor B, Alain T, Petruccelli LA, Hearnden J, Goncalves C, Grotegut S, Spruck CH, Furic L et al. (2015) Phosphorylation of eIF4E promotes EMT and metastasis via translational control of SNAIL and MMP‐3. Oncogene 34, 2032–2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Yuan X, Wu H, Bu H, Zheng P, Zhou J & Zhang H (2019) Design, synthesis and biological evaluation of pyridone–aminal derivatives as MNK1/2 inhibitors. Bioorgan Med Chem 27, 1211–1225. [DOI] [PubMed] [Google Scholar]
  • 116. Hou J, Lam F, Proud C & Wang S (2012) Targeting Mnks for cancer therapy. Oncotarget 3, 118–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Doehn U, Hauge C, Frank SR, Jensen CJ, Duda K, Nielsen JV, Cohen MS, Johansen JV, Winther BR, Lund LR et al. (2009) RSK is a principal effector of the RAS‐ERK pathway for eliciting a coordinate promotile/invasive gene program and phenotype in epithelial cells. Mol Cell 35, 511–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Ye Q, Wang X, Jin M, Wang M, Hu Y, Yu S, Yang Y, Yang J & Cai J (2018) Effect of RSK4 on biological characteristics of colorectal cancer. World J Surg Oncol 16, 240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Sulzmaier FJ & Ramos JW (2013) RSK isoforms in cancer cell invasion and metastasis. Cancer Res 73, 6099–6105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Silvera D, Formenti SC & Schneider RJ (2010) Translational control in cancer. Nat Rev Cancer 10, 254–266. [DOI] [PubMed] [Google Scholar]
  • 121. Golob‐Schwarzl N, Schweiger C, Koller C, Krassnig S, Gogg‐Kamerer M, Gantenbein N, Toeglhofer AM, Wodlej C, Bergler H, Pertschy B et al. (2014) Separation of low and high grade colon and rectum carcinoma by eukaryotic translation initiation factors 1, 5 and 6. Oncotarget 5, 101224–101243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Haybaeck J, O’Connor T, Spilka R, Spizzo G, Ensinger C, Mikuz G, Brunhuber T, Vogetseder A, Theurl I, Salvenmoser W et al. (2010) Overexpression of p150, a part of the large subunit of the eukaryotic translation initiation factor 3, in colon cancer. Anticancer Res 30, 1047–1055. [PubMed] [Google Scholar]
  • 123. Li Z, Lin S, Jiang T, Wang J, Lu H, Tang H, Teng M & Fan J (2014) Overexpression of eIF3e is correlated with colon tumor development and poor prognosis. Int J Clin Exp Pathol 7, 6462–6474. [PMC free article] [PubMed] [Google Scholar]
  • 124. Consortium TC , Consortium TE , Tomlinson IP, Webb E, Carvajal‐Carmona L, Broderick P, Howarth K, Pittman AM, Spain S, Lubbe S et al. (2008) A genome‐wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat Genet 40, 623–630. [DOI] [PubMed] [Google Scholar]
  • 125. Benedetti AD & Graff JR (2004) eIF‐4E expression and its role in malignancies and metastases. Oncogene 23, 3189–3199. [DOI] [PubMed] [Google Scholar]
  • 126. Rosenwald IB, Rhoads DB, Callanan LD, Isselbacher KJ & Schmidt EV (1993) Increased expression of eukaryotic translation initiation factors eIF‐4E and eIF‐2 alpha in response to growth induction by c‐myc. Proc Natl Acad Sci USA 90, 6175–6178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Gingras AC, Raught B, Gygi SP, Niedzwiecka A, Miron M, Burley SK, Polakiewicz RD, Wyslouch‐Cieszynska A, Aebersold R & Sonenberg N (2001) Hierarchical phosphorylation of the translation inhibitor 4E‐BP1. Gene Dev 15, 2852–2864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Chen Y, Wang J, Fan H, Xie J, Xu L & Zhou B (2017) Phosphorylated 4E‐BP1 is associated with tumor progression and adverse prognosis in colorectal cancer. Neoplasma 64, 787–794. [DOI] [PubMed] [Google Scholar]
  • 129. Shen P, Reineke LC, Knutsen E, Chen M, Pichler M, Ling H & Calin GA (2018) Metformin blocks MYC protein synthesis in colorectal cancer via mTOR‐4EBP‐eIF4E and MNK1‐eIF4G‐eIF4E signaling. Mol Oncol. 12, 1856–1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Rosenwald IB, Lazaris‐Karatzas A, Sonenberg N & Schmidt EV (1993) Elevated levels of cyclin D1 protein in response to increased expression of eukaryotic initiation factor 4E. Mol Cell Biol 13, 7358–7363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Shantz LM & Pegg AE (1999) Translational regulation of ornithine decarboxylase and other enzymes of the polyamine pathway. Int J Biochem Cell Biol 31, 107–122. [DOI] [PubMed] [Google Scholar]
  • 132. Kevil CG, Benedetti AD, Payne DK, Coe LL, Laroux FS & Alexander JS (1996) Translational regulation of vascular permeability factor by eukaryotic initiation factor 4E: implications for tumor angiogenesis. Int J Cancer 65, 785–790. [DOI] [PubMed] [Google Scholar]
  • 133. Fan S, Ramalingam SS, Kauh J, Xu Z, Khuri FR & Sun S‐Y (2009) Phosphorylated eukaryotic translation initiation factor 4 (eIF4E) is elevated in human cancer tissues. Cancer Biol Ther 8, 1463–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Rosenwald IB, Chen J‐J, Wang S, Savas L, London IM & Pullman J (1999) Upregulation of protein synthesis initiation factor eIF‐4E is an early event during colon carcinogenesis. Oncogene 18, 2507–2517. [DOI] [PubMed] [Google Scholar]
  • 135. Berkel HJ, Turbat‐Herrera EA, Shi R & de Benedetti A (2001) Expression of the translation initiation factor eIF4E in the polyp‐cancer sequence in the colon. Cancer Epidemiol Biomarkers Prev 10, 663–666. [PubMed] [Google Scholar]
  • 136. Gao M, Zhang X, Li D, He P, Tian W & Zeng B (2014) Expression analysis and clinical significance of eIF4E, VEGF‐C, E‐cadherin and MMP‐2 in colorectal adenocarcinoma. Oncotarget 5, 85502–85514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Xu T, Zong Y, Peng L, Kong S, Zhou M, Zou J, Liu J, Miao R, Sun X & Li L (2016) Overexpression of eIF4E in colorectal cancer patients is associated with liver metastasis. Oncotargets Ther 9, 815–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Xi C, Wang L, Yu J, Ye H, Cao L & Gong Z (2018) Inhibition of eukaryotic translation initiation factor 4E is effective against chemo‐resistance in colon and cervical cancer. Biochem Biophys Res Commun 503, 2286–2292. [DOI] [PubMed] [Google Scholar]
  • 139. Chen Y‐T, Tsai H‐P, Wu C‐C, Wang J‐Y & Chai C‐Y (2017) Eukaryotic translation initiation factor 4E (eIF‐4E) expressions are associated with poor prognosis in colorectal adenocarcinoma. Pathol Res Pract 213, 490–495. [DOI] [PubMed] [Google Scholar]
  • 140. Wolfe AL, Singh K, Zhong Y, Drewe P, Rajasekhar VK, Sanghvi VR, Mavrakis KJ, Jiang M, Roderick JE, der Meulen JV et al. (2014) RNA G‐quadruplexes cause eIF4Adependent oncogene translation in cancer. Nature 513, 65–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. García‐García C, Frieda KL, Feoktistova K, Fraser CS & Block SM (2015) Factor‐dependent processivity in human eIF4A DEAD‐box helicase. Science 348, 1486–1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Raza F, Waldron JA & Quesne JL (2015) Translational dysregulation in cancer: eIF4A isoforms and sequence determinants of eIF4A dependence. Biochem Soc Trans 43, 1227–1233. [DOI] [PubMed] [Google Scholar]
  • 143. Parsyan A, Svitkin Y, Shahbazian D, Gkogkas C, Lasko P, Merrick WC & Sonenberg N (2011) mRNA helicases: the tacticians of translational control. Nat Rev Mol Cell Biol 12, 235–245. [DOI] [PubMed] [Google Scholar]
  • 144. Rubio CA, Weisburd B, Holderfield M, Arias C, Fang E, DeRisi JL & Fanidi A (2014) Transcriptome‐wide characterization of the eIF4A signature highlights plasticity in translation regulation. Genome Biol 15, 476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Wang S, Darini C, Désaubry L & Koromilas AE (2016) STAT1 promotes KRAS colon tumor growth and susceptibility to pharmacological inhibition of translation initiation factor eIF4A. Mol Cancer Ther 15, 3055–3063. [DOI] [PubMed] [Google Scholar]
  • 146. Chen Z‐H, Qi J‐J, Wu Q‐N, Lu J‐H, Liu Z‐X, Wang Y, Hu P‐S, Li T, Lin J‐F, Wu X‐Y et al. (2019) Eukaryotic initiation factor 4A2 promotes experimental metastasis and oxaliplatin resistance in colorectal cancer. J Exp Clin Canc Res 38, 196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Peters TL, Tillotson J, Yeomans AM, Wilmore S, Jiménez‐Romero C, Amador LA, Amin AD, Li L, Pongtornpipat P, Zerio CJ et al. (2018) Target‐based screening against eIF4A1 reveals the marine natural compound elatol as a novel inhibitor of translation initiation with in vivo anti‐tumor activity. Clin Cancer Res 24, 4256–4270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Rosenwald IB, Wang S, Savas L, Woda B & Pullman J (2003) Expression of translation initiation factor eIF‐2α is increased in benign and malignant melanocytic and colonic epithelial neoplasms. Cancer 98, 1080–1088. [DOI] [PubMed] [Google Scholar]
  • 149. Koromilas AE (2015) Roles of the translation initiation factor eIF2α serine 51 phosphorylation in cancer formation and treatment. Biochim Biophys Acta 1849, 871–880. [DOI] [PubMed] [Google Scholar]
  • 150. Lobo MVT, Martín ME, Pérez MI, Alonso FJM, Redondo C, Álvarez MI & Salinas M (2000) Levels, phosphorylation status and cellular localization of translational factor EIF2 in gastrointestinal carcinomas. Histochem J 32, 139–150. [DOI] [PubMed] [Google Scholar]
  • 151. Kim SH, Gunnery S, Choe JK & Mathews MB (2002) Neoplastic progression in melanoma and colon cancer is associated with increased expression and activity of the interferon‐inducible protein kinase, PKR. Oncogene 21, 8741–8748. [DOI] [PubMed] [Google Scholar]
  • 152. Walter P & Ron D (2011) The unfolded protein response: from stress pathway to homeostatic regulation. Science 334, 1081–1086. [DOI] [PubMed] [Google Scholar]
  • 153. Dong J, Qiu H, Garcia‐Barrio M, Anderson J & Hinnebusch AG (2000) Uncharged tRNA activates GCN2 by displacing the protein kinase moiety from a bipartite tRNA‐binding domain. Mol Cell 6, 269–279. [DOI] [PubMed] [Google Scholar]
  • 154. Chu J, Cargnello M, Topisirovic I & Pelletier J (2016) Translation initiation factors: reprogramming protein synthesis in cancer. Trends Cell Biol 26, 918–933. [DOI] [PubMed] [Google Scholar]
  • 155. Nguyen HG, Conn CS, Kye Y, Xue L, Forester CM, Cowan JE, Hsieh AC, Cunningham JT, Truillet C, Tameire F et al. (2018) Development of a stress response therapy targeting aggressive prostate cancer. Sci Transl Med 10, eaar2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Jewer M, Lee L, Leibovitch M, Zhang G, Liu J, Findlay SD, Vincent KM, Tandoc K, Dieters‐Castator D, Quail DF et al. (2020) Translational control of breast cancer plasticity. Nat Commun 11, 2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Wang M & Kaufman RJ (2014) The impact of the endoplasmic reticulum protein‐folding environment on cancer development. Nat Rev Cancer 14, 581–597. [DOI] [PubMed] [Google Scholar]
  • 158. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, Yun C, Popko B, Paules R et al. (2003) An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 11, 619–633. [DOI] [PubMed] [Google Scholar]
  • 159. Heijmans J, van Lidth de Jeude JF, Koo B‐K, Rosekrans SL, Wielenga MCB, van de Wetering M, Ferrante M, Lee AS, Onderwater JJM, Paton JC et al. (2013) ER stress causes rapid loss of intestinal epithelial stemness through activation of the unfolded protein response. Cell Rep 3, 1128–1139. [DOI] [PubMed] [Google Scholar]
  • 160. Shi Z, Yu X, Yuan M, Lv W, Feng T, Bai R & Zhong H (2019) Activation of the PERK‐ATF4 pathway promotes chemo‐resistance in colon cancer cells. Sci Rep 9, 3210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Chern Y‐J, Wong JCT, Cheng GSW, Yu A, Yin Y, Schaeffer DF, Kennecke HF, Morin G & Tai IT (2019) The interaction between SPARC and GRP78 interferes with ER stress signaling and potentiates apoptosis via PERK/eIF2α and IRE1α/XBP‐1 in colorectal cancer. Cell Death Dis 10, 504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Hart LS, Cunningham JT, Datta T, Dey S, Tameire F, Lehman SL, Qiu B, Zhang H, Cerniglia G, Bi M et al. (2012) ER stress–mediated autophagy promotes Myc‐dependent transformation and tumor growth. J Clin Invest 122, 4621–4634. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Febs Journal are provided here courtesy of Wiley

RESOURCES