Skip to main content
RNA Biology logoLink to RNA Biology
. 2013 Jan 1;10(1):121–132. doi: 10.4161/rna.23312

DEAD box RNA helicase functions in cancer

Frances V Fuller-Pace 1,
PMCID: PMC3590229  PMID: 23353573

Abstract

Members of the DEAD box family of RNA helicases are known to be involved in most cellular processes that require manipulation of RNA structure and, in many cases, exhibit other functions in addition to their established ATP-dependent RNA helicase activities. They thus play critical roles in cellular metabolism and in many cases have been implicated in cellular proliferation and/or neoplastic transformation. These proteins generally act as components of multi-protein complexes; therefore their precise role is likely to be influenced by their interacting partners and to be highly context-dependent. This may also provide an explanation for the sometimes conflicting reports suggesting that DEAD box proteins have both pro- and anti-proliferative roles in cancer.

Keywords: DEAD box, multifunctional RNA helicases, cancer, oncogene, tumor suppressor

Introduction

DEAD box RNA helicases play key, and often essential, roles in RNA metabolism and generally function as components of large multi-protein complexes. Members of this family share a conserved core containing nine conserved motifs, including the characteristic D-E-A-D motif; these motifs confer the ATP hydrolysis and RNA unwinding activities that have established them as RNA helicases. However, it is clear that many DEAD box proteins are multifunctional and have additional diverse roles that are conferred by their distinct N- and C-terminal domains and dependent on their interactions with partner proteins. There have been numerous reports indicating that DEAD box proteins are involved in processes that are key to cellular proliferation and/or neoplastic transformation; therefore, it is not surprising that deregulation of expression or function of these proteins has been implicated in cancer development or progression.1

In this review I shall focus on the DEAD box proteins DDX1, DDX3, DDX6, DDX5 and DDX17, proteins for which there is a significant body of evidence for their involvement in cancer development. Interestingly, all of these proteins have been suggested to have both oncogenic and tumor-suppressive roles in cancer and there have been often conflicting reports on their role in cancer development. However the data presented to date suggest that their precise function may be highly tumor- and/or context- dependent, and may be influenced by their interacting partners. I shall review the multiple functions that have been attributed to these proteins and discuss how these can act to promote or inhibit tumor development in different contexts. These are summarized in Table 1.

Table 1. The multiple functions attributed to the DEAD box proteins DDX1, DDX3, DDX6, DDX5 and DDX17, and their potential importance in tumor development.

Protein* Function Targets / Context Importance in Cancer
DDX1
Viral replication, nuclear export of viral RNAs
Export of HIV RNA, JC virus and HCV replication14
Co-amplified with MYCN in neuroblastoma and retinoblastoma2-6
Overexpressed in breast cancer7 and testicular carcinoma22
Cell migration and anchorage- independent growth19,22
Most reports suggest oncogenic properties but some imply tumor-suppressive roles in some contexts.3-13
3′end processing of mRNAs
Associated with cleavage stimulating factor in HeLa cells16
RNA Transport /
RNA clearance
Associated with RNA transporting granules (with DDX3) in neuronal dendrites15
RNA clearance from DNA strand break sites in HeLa cells17
Cytoskeletal reorganization /
cell migration
Cellular elongation and migration in monocytes19
Anchorage independent growth in testicular germ cell tumor cell line22
Transcription
Activation of stem cell-associated genes (e.g., Cyclin D2) in testicular germ cell tumors22
DDX3, DDX3X, DBX
(Ded1p)
Viral replication, nuclear export of viral RNAs
HIV, HBV, HCV: potential drug target23,24
Both oncogenic and tumor suppressor roles suggested in HCC and breast cancer42-51
Overexpression in HCC,43 promotes anchorage independent growth43 and cell cycle progression,44 inhibits apoptosis46,47
Reduced expression in some HCC,49 inhibits cell cycle progression through activation of p21WAF1 transcription in HCC and lung cancer40,49,50
Stimulates migration / invasion42
Context-dependent regulation of RNA export and translation could have important implications in tumor cell growth.
Pre-mRNA splicing
DDX3 and the yeast homolog, Ded1p, associated with mRNPs and spliceosomes but no direct evidence for functional role26-28
RNA export
Export of unspliced or partially spliced HIV transcripts29
Associated with RNA transporting granules (with DDX1) in dendrites15 and with the mRNP export receptor TAP30
Translation
Can both promote and repress translation initiation: associates with P-bodies, ATP hydrolysis allows release from P-bodies and translation initation32-37
Transcription
Activation of IFNβ, p21WAF1, Snail; inhibition of E-cadherin transcription38-42,60
EMT, cell migration / invasion
DDX3 overexpression induces EMT and motility/invasiveness in MCF10 breast cancer cells42
DDX6, Rck/p54
(Xp54; p47; ME31B; Dhh1p)
mRNP export, translational repression in development
Associated with cytoplasmic mRNP storage particles and translational repression in Drosophila and Xenopus oocytes58,65; Developmental roles in many species57-62
Overexpression in several cancers53-56
Stimulation of cell cycle progression and proliferation70-74
Aberrant regulation of RNA export, RNA degradation and translation could have important implications in tumor cell growth
Most reports suggest oncogenic properties70-74but some imply tumor-suppressive roles in certain contexts69,75
RNA degradation / Translation repression
Associated with P-bodies and translational repression in HeLa cells66,70 and yeast69
Associated with RISC in HeLa cells, important for miRNA induced translation repression67
Dhh1p associated with decapping and deadenylase complexes in yeast,68,69 can promote mRNA degradation71
Cell cycle progression / proliferation
Dhh1p important for recovery from DNA damage-induced G1/S cell cycle arrest in yeast
DDX6 required for S-phase progression and cell viability in HeLa and LoVo cells,73,74 and proliferation in xenograft models74 (siRNA studies)
In contrast, DDX6 or Dhh1p overexpression inhibits cell growth75,69
DDX5, p68
DDX17,
p72
Transcription
DDX5 coactivates ERα97, p53,100 MyoD,99 AR,84 Runx298, NF-κB87
DDX5 required for p53 DNA damage response100 and activation of selective p53 target genes101
DDX17 coactivates ERα and required for activation of ERα target genes85
DDX5 / DDX17 activate transcription of both pro- proliferation and anti-proliferative genes,84-104 regulate promoter switching110
DDX5 / DDX17 aberrantly expressed/modified in a range of cancers83-89
DDX5 / DDX17 coactivate transcription factors that play important roles in tumor development84-110
DDX5 activates transcription of genes important for DNA replication.
DDX5 stimulates EMT and DDX5 / DDX17 modulate cell migration
DDX5 selectively activates p53-dependent p21WAF1 induction; influences cell cycle arrest/ apoptosis decision101
Deregulation of splicing/alternative splicing and miRNA biogenesis play important roles in tumor development
Several reports suggest oncogenic roles for DDX5 / DDX17, but also evidence for context-dependent tumor suppressor roles1,84-108
Pre-mRNA splicing / alternative splicing
DDX5 / DDX17 associated with spliceosomes, required for pre-mRNA splicing111-113
Alternative splicing and exon skipping114-118
RNA clearance, nuclear export
DDX5 implicated in RNA clearance from chromatin and export18,31
miRNA biogenesis
DDX5 /DDX17 components of Drosha complex, implicated in miRNA processing of subsets of miRNAs119-124
Ribosome biogenesis
DDX5 / DDX17 implicated in rRNA processing (knockout mice)79
DDX5 / DDX17 required for ribosome biogenesis in HeLa cells94
EMT, cell migration DDX5 regulates PDGF-induced EMT88
DDX5 / DDX17 modulate cell migration through NFAT5 regulation108
*

Alternative names, including homologs in species other than human, are also shown. Homologs are shown in brackets.

DDX1

DDX1 amplification and potential role in cancer

One of the first indications that DDX1 may be involved in tumor development came from reports that it is co-amplified with the MYCN gene in retinoblastomas and neuroblastomas.2-5 The finding that co-amplification of DDX1 and MYCN was more frequent in higher stages of neuroblastoma and was associated with a significant reduction in disease-free survival compared with those with only MYCN amplification3,6 suggested that DDX1 has oncogenic properties. This idea is supported by a more recent study of breast cancer gene expression and tissue microarrays, which showed that DDX1 RNA overexpression and elevated cytoplasmic DDX1 protein are associated with early recurrence and suggested that DDX1 may be an independent prognostic marker for early recurrence in breast cancer.7

However, there have been several conflicting reports concerning the role of DDX1 in tumor development. For example, in one report high DDX1 expression in neuroblastoma was associated with better survival,8 while a recent study showed that DDX1 expression was associated with improved local relapse-free-, distant metastasis-free- and overall survival in patients diagnosed with early-stage node-negative breast cancer,9 suggesting a possible tumor suppressor role for DDX1. However, De Preter and colleagues found no evidence of any effect of DDX1 amplification on prognosis of patients with MYCH-amplified neuroblastomas,10,11 while other reports suggested that the prognostic effect of DDX1 amplification/overexpression on MYCN is different between different subgroups,12,13 providing a possible explanation for the different results obtained from the various studies. Although most reports indicate an oncogenic role for DDX1 in tumor development, much of the evidence is circumstantial rather than mechanistic. Therefore, its precise function may depend both on the cancer type (or even subtype), treatment administered to patients, as well as context: i.e., the expression of other factors that may influence both DDX1 function and, independently, the treatment chosen for the specific cancer (e.g., Estrogen Receptor status in breast cancer).

DDX1 functions

Apart from the well-documented functions in the replication of several viruses, most notably HIV (reviewed in ref. 14), DDX1, like many other DEAD box proteins, is found to be a component of several cellular protein and ribonucleoprotein complexes.

3′ end processing of pre-mRNAs

DDX1 is predominantly nuclear in many cells in which the DDX1 gene is not amplified, while it is present in both the nucleus and cytoplasm in cells in which the gene is amplified/overexpressed (reviewed in ref. 14). However it has also been reported in the cytoplasm in some cell lines (e.g., neurons, see ref. 15 and below). Apart from showing a general punctate nucleoplasmic distribution, DDX1 is also found in discrete nuclear foci and is associated with the cleavage stimulation factor CstF-64, suggesting a role in 3′-end polyadenylation and cleavage of pre-mRNAs.16 Additionally DDX1 was also found to interact with poly(A) RNA and with heterogeneous nuclear ribonucleoprotein K (hnRNP K) a protein that is involved in multiple stages of mRNA biogenesis/metabolism including chromatin remodeling, transcription regulation, pre-mRNA processing, nuclear transport and translation. These findings are consistent with the idea that at least one of the functions of DDX1 is in 3′ end processing of pre-mRNAs; however to date there is no evidence for a direct role in this process.

RNA transport/clearance

DDX1, together with DDX3 and DDX5 (see below), was identified as a protein associated with kinesin KIF5 in an RNA-transporting granule15 in neuronal dendrites, suggesting a potential role in RNA transport. In this respect the association of DDX1 with hnRNP K would also support a role for DDX1 in RNA transport. Furthermore, a study demonstrating the re-distribution of DDX1 to ionizing radiation-induced foci and its co-localization with γ-H2AX and phosphorylated ATM, coupled with its ability to unwind DNA-RNA hybrids, suggest a possible role for DDX1 in RNA clearance from double strand break sites.17 This is interesting because a RNA clearance role has also been implicated for DDX5 (see below).18 Furthermore, the idea that DDX1 functions in RNA clearance from DNA double strand break sites implies a role in transcription-coupled repair, which may in turn have implications for DDX1 function in tumor development.

Cytoskeletal reorganization/cell migration

A potential role for DDX1 in cell motility has emerged from reports identifying DDX1 in (1) complexes with LRFN4, a neuronal transmembrane protein that is also expressed in monocytes and signals cytoskeletal reorganization and morphological cell elongation,19 and (2) in proteomic analysis of the pseudopodial fractions of invasive variants of Moloney sarcoma virus- transformed Madin-Darby canine kidney cells. Although they do not specifically demonstrate that DDX1 is functionally involved in cell motility these observations are important since cell migration and/or invasion are key steps in tumor invasion and metastasis.

Interestingly, in one of these reports,19 the complex containing DDX1 also contained several 14–3-3 proteins, which have been found to be involved in signal transduction, cell cycle control and apoptosis. Other studies also identified DDX1 as a component of 14–3-3-containing complexes20,21 consistent with the idea that DDX1 may play a part in multiple cellular processes that are implicated in tumor development or progression.

Transcription: Transcriptional activation of stem cell associated genes

One of the strongest pieces of evidence linking DDX1 with tumor development comes from a study showing that it is required for transcriptional activation of stem cell associated genes (including cyclin D2) that are implicated in testicular germ cell tumors.22 In this study, siRNA knockdown of DDX1 resulted in a striking inhibition in their anchorage-independent growth and an abrogation of their ability to form tumors in nude mice, indicating that DDX1 is critical for tumorigensis and highlighting potential mechanisms by which deregulation of DDX1 could play a crucial role in cancer development and/or progression.

DDX3

In recent years DDX3 has gained much attention due to its role in the replication of viruses that are of global health importance (including HIV, HBV, HCV) and there are major efforts aimed at developing strategies to target DDX3 therapeutically (reviewed in refs. 23,24), including, most recently, small molecule inhibitors that target the DDX3 RNA binding site as a potential anti-HIV therapy.23 However, numerous studies have demonstrated that DDX3 and its yeast homolog (Ded1p) function in multiple cellular processes involved in the regulation of gene expression (reviewed in refs. 23,25). These include transcription, pre-mRNA splicing and mRNA export and translation, functions that are often associated with several other DEAD box proteins. Additionally, DDX3 has also been implicated in cell cycle regulation and apoptosis, consistent with it being important in cancer development. Interestingly, however, as appears to be the case for the other DEAD box proteins discussed here, there are reports suggesting both oncogenic and tumor suppressor functions for DDX3.

DDX3 functions

DDX3 (also known as DDX3X, DBX) has been variously described to be a nuclear protein, a cytoplasmic protein, and as a protein that shuttles between the nucleus and the cytoplasm, consistent with its described nuclear and cytoplasmic functions.

Pre-mRNA splicing/RNA export

Although initial studies suggested that the yeast Ded1p was involved in pre-mRNA splicing, its role in this process remains unclear. Nevertheless, Ded1p and the human DDX3 were found to associate with mRNPs and spliceosomes,26-28 but whether DDX3/Ded1p is actively involved in splicing or whether it functions in export of spliced mRNAs remains to be fully elucidated.

DDX3 was first shown to be important for the export of unspliced or partially spliced HIV transcripts29 and subsequently was found to associate with the major nuclear mRNA export receptor, Tip-associated protein (TAP), with mRNPs30 and with RNA transporting granules,15 shuttling from the nucleus to the cytoplasm. These studies suggest that DDX3 is unlikely to be involved in general mRNA export; instead it may be involved in the export of viral RNAs and specific subsets of cellular mRNAs and may subsequently also be important in regulating the translation of these mRNAs. Interestingly, DDX3 was recently shown to interact with another DEAD box helicase, DDX5, (see below) and suggested to influence DDX5 shuttling during mRNP export.31

Translation

Perhaps the best studied function for DDX3, and the yeast Ded1p, is their role in translation. Work from several groups showed that the Ded1p promotes translation initiation32,33 and, indeed, many studies have reported interactions between DDX3/Ded1p and proteins involved in translation initiation (reviewed in ref. 23). However there have been other conflicting results: for example, one study reported that DDX3 represses cap-dependent translation by binding and inhibiting eIF4E34; others suggested that DDX3 promotes translation of specific mRNAs that contain secondary structures in their 5′UTR through interaction with eIF4F.30,35 Yet another study suggested that the yeast Ded1p can both repress and promote translation through its association with P-bodies (Processing bodies).36 This was confirmed by data showing that Ded1p interacts with eIF4G to assemble a translationally repressed Ded1p-mRNA-eIF4F complex in P-bodies but subsequently, upon ATP hydrolysis, resolves the “stalled” mRNP and allows its exit from the P-bodies for translation initiation thus controlling the release (and translation) of specific mRNAs in an ATP-dependent manner.37 Interestingly, the inhibitory effect on translation initiation through eIF4E inhibition did not appear to require helicase activity34 while the resolving of the stalled mRNP requires ATP hydrolysis and, presumably, RNA unwinding.37 Thus, the precise function of DDX3/Ded1p in translation might also be modulated by other factors or conditions in the cell (e.g., stress conditions). This could have important implications in cancer and might, at least in part, explain the different reports suggesting both oncogenic and tumor suppressor roles for DDX3 (see below).

Transcription: Transcriptional regulation of genes important in cancer

Several studies from different groups have reported that DDX3 can regulate transcription of genes that are important in cancer. These include activation of the Interferon-β (IFNβ),38,39 the cell cycle arrest gene p21waf140 and the transcription factor Snail41 promoters, and repression of the E-cadherin promoter.42 Chromatin Immunoprecipitation assays demonstrated that DDX3 is recruited to the IFNβ, E-cadherin and the p21waf1 promoters,38-40,42 suggesting that DDX3 functions in transcriptional initiation. Similarly, the DDX3-dependent effect on Snail transcription in the presence of histone deacetylase (HDAC) inhibitors41 is consistent with DDX3 being involved in initiation of Snail transcription.

Interestingly, DDX3 appears to work in subtly different ways in regulating transcription of these genes. For example activation of the IFNβ promoter appears to be independent of ATPase or RNA helicase activity,38,39 while activation of the p21waf1 promoter requires ATPase, but not RNA helicase activity,40 suggesting promoter- or context-dependent effects. Interestingly similar promoter-dependent differences have been observed for DDX5 (see below).

In terms of the role of DDX3 in cancer, stimulation of Snail41 and repression of E-cadherin42 would be consistent with DDX3 promoting epithelial-mesenchymal transition (EMT) since these genes play key roles in this process. EMT is one of the pre-requisites for cancer invasion and metastasis, suggesting an oncogenic role for DDX3. In contrast, its activation of the cell cycle arrest gene p21waf140 and its activation of IFNβ38,39 (which has been shown to inhibit angiogenesis, a process that is critical for cancer growth) would imply a growth-, or tumor-suppressor, role for DDX3. Thus, as discussed below, the precise role of DDX3 in cancer may be tumor and/or context-dependent and is also likely to be dependent on the availability of other factors that regulate transcription.

DDX3 in cancer: oncogene or tumor suppressor?

DDX3 as an oncogene

The first suggestion that DDX3 can act as an oncogene came from a study of hepatocellular carcinoma (HCC), which reported overexpression of DDX3 mRNA in a panel of HCC tissues and showed that ectopic expression of DDX3 stimulated anchorage-independent growth of HCC cells, a hallmark of cell transformation.43 These findings are supported by data from two reports showing that (1) overexpression of DDX3 repressed E-cadherin expression and increased motility in breast cancer cell lines42; and (2) was required for Snail expression, an inducer of EMT.41

Additionally, DDX3 has been shown to promote translation initiation of cyclin E1 thus facilitating G1/S transition and promoting cell growth44; this is consistent with an earlier report that Hamster tsEt24 cells, which harbor a temperature-sensitive mutation in DDX3, exhibit G1 arrest at the non-permissive temperature.45 Other studies have suggested that DDX3 has an anti-apoptotic function. DDX3 was found to be important in blocking tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated apoptosis by associating with the TRAIL receptor 2 (TRAIL-R2/DR5) and blocking apoptotic signaling46; this finding was supported by data showing that DDX3 siRNA knockdown increased TRAIL-R2- mediated apoptosis47 and that breast cancer cell lines expressing low levels of DDX3 were more sensitive to antibodies targeting this receptor.48

Taken together the above data support the notion that DDX3 can enhance cell proliferation through cell cycle regulation, inhibit apoptosis, and promote cell transformation and migration. These are all consistent with DDX3 having a tumor-promoting or oncogenic role in the cell.

DDX3 as a tumor suppressor

However, there is also evidence that, at least in some contexts, DDX3 has a tumor suppressor function. In contrast to the first report of DDX3 overexpression in HCC,43 a subsequent study showed a reduction of DDX3 expression in HCC from hepatitis B-positive but not hepatitis C-positive patients.49 Moreover, the latter study also showed that siRNA knockdown of DDX3 in the non-transformed mouse cell line NIH3T3 results in enhanced cell proliferation and premature entry into S-phase, concomitant with downregulation of p21waf1 and upregulation of cyclin D1; these data clearly contrast with those indicating that DDX3 can promote G1/S transition and cell growth- see above.44,45 Additionally, Chang and colleagues49 reported that constitutive DDX3 siRNA knockdown in NIH3T3 cells results in an increase in ras-induced anchorage-independent growth and resistance to apoptosis induced by serum depletion; these data again conflict with those from the reports by Huang, Sun and colleagues.43,47

In another study, ectopic expression of DDX3 was found to inhibit cell growth, as seen by colony formation assays in a range of tumor cells, in a p21-dependent manner through transcriptional activation of the p21waf1 promoter.40 Similarly, in a separate report, DDX3 was shown to synergistically stimulate p53-dependent transcription of p21waf1; moreover DDX3 was positively associated with p21 expression in lung cancer and reduced expression of DDX3/p53 and p21 were found to associate with poor relapse-free survival.50 Both these studies support a tumor suppressor role for DDX3, through activation of p21 expression. These data contrast with those from Botlagunta and colleagues,42 which showed that DDX3 has no effect on p21 expression in breast cancer MCF-7 cells and high DDX3 levels were associated with low p21 levels in aggressive breast cancers.

To further complicate the picture, DDX3 was found to inhibit cell growth in the human hepatocellular carcinoma cell line HuH-7 through repressing cap-dependent translation by binding and inhibiting the translation initiation factor eIF4E.34 This conflicts with the data from Lai and colleagues44 and is consistent with observations that DDX3 can both stimulate and inhibit translation in a context-dependent manner.

It is not clear why alterations in DDX3 levels produce such opposing tumor-suppressing and proliferation- or tumor-promoting effects in different cell lines, although the effect may depend on the expression of other cellular factors in the different systems and/or the model systems/genes under study. In this respect, alterations in nucleo-cytoplasmic shuttling and therefore the cellular localization of DDX3 may be important since it could have profound effects on its function, depending on context. The reasons behind the discrepancies concerning DDX3 localization in the various reports remain unclear (reviewed in ref. 23), although one study did show an alteration of DDX3 subcellular localization from the nucleus to the cytoplasm in a high percentage of cutaneous squamous cell carcinoma.40 This may also perhaps explain the conflicting reports showing an increase in DDX3 expression in some cases, and a decrease in others, being associated with cancer development and/or progression.

DDX6

DDX6 (also known as Rck/p54) was first suggested to be involved in tumor development through its cloning as a gene found at a chromosomal 11q23 breakpoint in the RC-K8 B-cell lymphoma line, when it was shown to be highly homologous to the Drosophila DEAD box gene ME31B.51,52 Subsequent reports demonstrated that the DDX6 protein is overexpressed in glioblastoma, rhabdosarcoma and lung cancer cell lines as compared with normal tissue,53 in colorectal tumors54,55 and in cases of HCV-related chronic hepatitis and hepatocellular carcinoma.56 All these implied an involvement of DDX6 in cancer development, although the precise mechanism of its contribution to cancer was unclear at the time.

DDX6 functions

Developmental functions have been suggested for DDX6 homologs in several organisms including the Drosophila ME31B,57,58 the Schizosaccharomyces pombe Ste13,59 the Xenopus Xp54,60 the Caenorhabditis elegans Cgh-161 and the murine Rck/p54.62 Further functional studies on these homologs have demonstrated that DDX6-like proteins play important roles in several cellular processes including mRNP assembly/export, RNA degradation and translational regulation (reviewed in ref. 63).

mRNP assembly and export: association with P-bodies, RNA degradation and translational repression

The Xenopus Xp54 was found to shuttle between the nucleus and cytoplasm in Xenopus oocytes; it was shown to be involved in the export of maternal mRNAs and to localize to mRNP storage particles in the cytoplasm.64 Similarly the Drosophila ME31B58 and the Clam homolog p4765 were shown to associate with RNP complexes and to repress translation of oocyte mRNAs. A more recent study using both the Xenopus Xp54 and human DDX6 demonstrated that RNA helicase activity is required for translation repression.66 However, while DDX6 helicase activity was required for P-body assembly in HeLa cells the C-terminal domain (outside the helicase core) was sufficient for translational repression66; and suggested that ATP-dependent conformational changes in DDX6 and modulation of its binding to protein partners may be crucial in regulating translation. This implies that DDX6 is multifunctional and has helicase-dependent and -independent functions that depend on interaction with other partners. This is reminiscent of the findings with DDX3 and DDX5, and may be a common property of many DEAD box proteins.

Interestingly, DDX6 has also been found to interact with the Argonaute proteins Ago1 and Ago2, associate with the RNA-induced silencing complex (RISC) and to be important for miRNA-induced, but not siRNA-induced, translational repression67 suggesting a further mechanism by which DDX6 may repress translation.

Other studies have shown that Dhh1p, the Saccahromyces cerevisiae homolog of DDX6 functions in mRNA decapping and is associated with both decapping and deadenylase complexes68,69 providing a mechanism by which it can regulate translation of mRNAs through RNA degradation. These findings are supported by recent data, which shows that the human DDX6 is bound to repressed mRNAs at multiple points and proposes that this binding aids their localization to P-bodies and the recruitment of decapping complexes.70 Interestingly, Dhh1p was similarly shown to promote degradation of a specific mRNA (porin mRNA) in yeast through interaction via its C-terminal domain,71 again suggesting that this function is helicase-independent (see above).

Regulation of cell cycle progression and proliferation: DDX6 as an oncogene

Several studies, with both the human DDX6 and the yeast Dhh1p, have indicated that this protein acts as a positive regulator of cell cycle progression: Dhh1p has been shown to be important for recovery from DNA-damage-dependent G1/S cell cycle arrest,72 while two separate studies demonstrated that siRNA knockdown of DDX6 resulted in S-phase cell cycle arrest.73,74

Furthermore, these studies showed that siRNA knockdown of DDX6 resulted in a reduction in cell viability,73 as well as an increase in cell apoptosis and a reduction in their ability to form tumors in xenograft models.74 Additionally, overexpression of DDX6 was shown to increase expression of the oncogene c-Myc, consistent with the idea that it would have a positive effect on cell proliferation.55

Taken together, these findings suggest that DDX6 homologs play an important, evolutionarily conserved, positive role in cell cycle progression and proliferation, perhaps through regulating translation of specific mRNAs that play key roles in cell cycle progression. This would be consistent with the idea that DDX6 can act as an oncogene and is supported by the many reports showing that it is overexpressed in a wide range of cancers.53-56

Growth inhibitory/tumor suppressor roles for DDX6

In contrast to the above, there has been one report that overexpression of DDX6 results in an inhibition of cell growth and a reduction in anchorage-independent growth, a hallmark of malignant transformation.75 Furthermore, overexpression of Dhh1p was found to inhibit cell growth in yeast, suggested to be through general repression of translation.69 While these results suggest that DDX6 homologs in these contexts have a negative role on cell proliferation and conflict with those described above, the discrepancy could be due to the fact that most of the experiments showing a positive effect of DDX6 on cell proliferation used siRNA depletion of endogenous DDX6 while those showing a negative effect on proliferation relied on ectopic overexpression. Perhaps having the correct physiological amount of DDX6 in cells is critical and the precise function of DDX6 will depend on the levels of the protein in the cell as well as the availability of interacting partners.

DDX5 and DDX17

DDX5 (also known as p68) is one of the prototypic members of the DEAD box family of RNA helicases.76 DDX5 and the highly related DDX17 (p72)77 have been shown to function in multiple processes in the cell, including transcription, pre-mRNA and -rRNA processing, alternative splicing and miRNA processing, and there have been several reports showing that their expression is deregulated in a range of cancers. Furthermore, both DDX5 and DDX17 have been functionally implicated in tumor development: several reports have suggested oncogenic or pro-proliferation roles for these proteins while some have implied potential tumor suppressor roles (reviewed in ref. 78). Furthermore, it is clear that post-translational modification of DDX5 and DDX17 has profound effects on DDX5/DDX17 function and this may be critical to their role in cancer.

The DDX5 and DDX17 genes share a remarkable degree of homology.77 DDX5 and DDX17 function in multiple cellular processes; however, although they appear to share some functional overlap in some contexts, several reports have shown that they also have specific non-redundant functions (reviewed in ref. 78), perhaps most clearly demonstrated by the results from DDX5 and DDX17 knockout mice.79

Expression and modification of DDX5 and DDX17 in cancer

In the original paper describing the discovery of DDX5,81 the observation that exponentially growing cells appeared to have higher levels of the protein suggested that DDX5 may somehow be involved in cell proliferation. A subsequent report indicated that DDX5 expression was developmentally regulated and associated with organ maturation /differentiation but that, in cell lines, expression was induced by serum suggesting that the regulation of DDX5 expression is complex.80

The first report of aberrant DDX5 expression in cancer came from a study which showed that DDX5 was overexpressed and abnormally modified (by polyubiquitylation) in colorectal cancer.81 This was followed by several other reports showing DDX5, and in several cases also DDX17, overexpression in a range of cancers including colon,82,83 prostate,84 breast,85 cutaneous squamous cell carcinoma86 and glioma.87

Phosphorylation of DDX5, in particular on Tyrosine593, has been reported to be associated with cellular transformation and tumor development82 and to alter DDX5 localization and function. DDX5 phosphorylated on Y593 through the action of the Platelet-Derived Growth Factor (PDGF)-activated c-Abl kinase] was found to be exported from the nucleus to the cytoplasm and to facilitate β-catenin nuclear import.88 Furthermore it was shown to be important for PDGF-induced Epithelial-Mesenchymal Transition (EMT).88 Similarly PDGF-induced phosphorylation of DDX5 was found to be important for β-catenin nuclear translocation and activation in prostate cancer cells.89 Although some other studies reported that Y593 phosphorylation on DDX5 was not required for β-catenin nuclear localization,83,90 it is clear that phosphorylation by c-Abl affects DDX5 function, at least in certain contexts, as shown by its effects on induction of cyclin D1 and c-Myc expression,91 Snail192 and Androgen Receptor target genes84 (see below). Furthermore a double phosphorylation on Y593 and Y595 of DDX5 was found to impart resistance to apoptosis in gliobastoma cells treated with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL).93

Taken together these findings suggest that post-translation modification of DDX5 and DDX17 may play important roles in cancer development/progression and that DDX5/DDX17 have pro-proliferation/-transformation roles that can be modulated by post-translational modification. This idea is supported by reports showing that simultaneous depletion of both DDX5 and DDX17 results in an inhibition of both proliferation of cancer cells and of their ability to form tumors in xenograft mouse models,83,94 and that DDX5 depletion inhibited transformation of RasV12 transformed Arf-deficient cells in vitro and in vivo.95 It is also consistent with reports that DDX5 expression associates with higher grade in prostate and breast cancers,84,85 gliomas87 and metastatic cutaneous squamous cell carcinoma.86 Furthermore, a recent report demonstrated that DDX5 is amplified in a subset of breast cancers; in these cancers DDX5 is required for proliferation and inhibition of DDX5 sensitizes this subset of cancer cells to trastuzumab,96 suggesting that DDX5 is oncogenic in certain cancers and could even be a therapeutic target.

However, in the study of Estrogen Receptor α (ERα)-positive breast cancers which showed the association of DDX5 with higher grade and markers of poor prognosis, but not significantly with survival, DDX17 was associated with better survival85 suggesting that the role of DDX5 and DDX17 in cancer development and progression is more complex and is likely to be context-dependent, as discussed below.

DDX5 and DDX17 functions: oncogenes or tumor suppressors?

Regulation of Transcription

There is now a large body of evidence demonstrating that DDX5 and DDX17 act as coactivators of transcription factors that are themselves highly regulated and play important roles in cancer development. Much of the work to date in this area has been performed on DDX5, perhaps reflecting the higher availability of reagents; however it is clear that DDX17 also acts as a regulator of transcription, with some functions that are distinct from those of DDX5. Chromatin immunoprecipitation assays indicate that DDX5 and DDX17 are recruited to responsive promoters and act, at least in part, by modulating transcriptional initiation. Interestingly, DDX5/DDX17 RNA helicase activity appears to be required for transcriptional regulation in some, but not all, cases (reviewed in ref. 78), suggesting that the precise function of DDX5/DDX17 in transcription may be dependent on the promoter context and/or on other interacting factors.

Consistent with the idea that DDX5 has pro-proliferation or oncogenic roles in cancer, it has been shown to coactivate ERα,97 Androgen Receptor (AR),84 Runx2,98 the NF-κB transcription factor p50,87 and to upregulate expression of cyclin D1, c-Myc (consistent with β-catenin activation88,91 as well as genes required for DNA replication.96 Furthermore, DDX5 was found to activate transcription of the Snail1 gene by displacing histone deacetylase from the Snail1 promoter,92 consistent with an involvement in EMT. Interestingly Y593 phosphorylation was found to be important for this function, as previously shown for DDX5 activation of AR,84 underscoring the idea that phosphorylation of DDX5 can modulate its function.

On the other hand, anti-proliferative or tumor-suppressor functions have also been implied for DDX5 and DDX17. A pro-differentiation role was suggested by the finding that DDX5 and DDX17 coactivate the myogenic regulatory factor MyoD and are required for skeletal muscle cell differentiation.99 DDX5 has also been shown to coactivate the p53 tumor suppressor and to be required for the p53-dependent DNA damage response.100 However, such a role is likely to be context-dependent since a recent study demonstrated that while DDX5 is required for p53-dependent p21waf1 induction and cell cycle-arrest it is not required for apoptosis induction,101 suggesting that in certain cases DDX5 could have a pro-survival role. Furthermore, the finding that DDX5 induces expression of the cell cycle arrest gene p21waf1,101 and conversely cyclin D1 in a different context,91 suggests that DDX5, under different conditions, could have opposing effects on cell cycle progression. These findings underscore the idea that DDX5 function is highly dependent on context and presumably, post-translational modification. Additionally, a recent study found that DDX5 coactivates the vitamin D receptor and enhances its response to the activated vitamin D ligand, calcitrol, again suggesting an anti-proliferative role for DDX5 and proposing a possible p53-independent way of inducing p21 waf1 expression.102

Sumoylation and acetylation have also been shown to modulate DDX5/DDX17 function as transcriptional regulators.103-105 It is also clear that both DDX5 and DDX17 can act as transcriptional repressors,106,107 with sumoylation enhancing their repressor activity in some, but not all, contexts,103,104 implying further context- and possibly tumor-dependent regulation of their function.

A further complexity is provided by the recent finding that DDX5/DDX17 have dual and opposing roles in regulating the pro-migration transcription factor NFAT5: they enhance NFAT5 transcriptional activity and increase cell migration but they also modulate alternative splicing of NFAT5 leading to the inclusion of exon 5, which includes a translation termination codon and leads to nonsense-mediated decay.108 Thus DDX5/DDX17 can both enhance NFAT5 transcriptional activity and, by regulating its splicing, reduce its protein expression level.

Other functions for DDX5 and DDX17 in more global modulation of transcription have been described. These include: (1) transcriptional “insulation” by DDX5 through interaction with CTCF, a DNA binding protein involved in chromatin organization, to avoid inappropriate activation or repression of genes109; (2) Estrogen-regulated promoter switching by DDX5 and DDX17 to modulate ERα-mediated transcription110; and (3) RNA clearance from chromatin to allow gene deactivation and possibly RNA export.18,31 These would have important implications for context-dependent functions for DDX5/DDX17 and may, at least in part, explain the conflicting reports implying both oncogenic and tumor suppressor roles for these proteins.

Splicing/alternative splicing

DDX5 and DDX17 have been shown to be associated with the spliceosome and to function in pre-mRNA splicing.111-113 Perhaps more interesting in terms of their potential role in cancer development are the reports demonstrating that they can regulate alternative splicing. Of particular interest are the data showing that DDX5 and DDX17 can modulate alternative splicing that is regulated by steroid hormone receptors, with DDX5 enhancing AR-mediated exon skipping84 and DDX17 (but not DDX5) inhibiting ERα/ERβ-mediated exon skipping.114,115 Furthermore, DDX5 was found to modulate alternative splicing of the H-Ras proto-oncogene, enhancing skipping of the alternative D (IDX) exon and thus altering the balance in favor of the more oncogenic p21 H-ras isoform.116,117

Recently, in a mouse model of tumor progression, DDX5 and DDX17 were found to contribute to tumor cell migration and invasion by regulating alternative splicing of several DNA-binding factors, including the epigenetic chromatin regulator macroH2A1 histone (mH2A1).118 Interestingly either DDX5 or DDX17 depletion inhibited cell migration/invasion, with simultaneous depletion of both proteins having an enhanced effect.

This study also demonstrated that the mH2A1.1 and mH2A1.2 isoforms of mH2A have opposing effects on the transcription of the redox metabolism gene, superoxide dismutase SOD3, which inhibits cell migration. DDX5 and DDX17 favored expression of mH2A1.2, which inhibits expression of SOD3. Thus, through modulation of mH2A1 alternative splicing, DDX5/DDX17 could regulate redox metabolism and cell migration/invasion, critical factors in cancer progression and metastasis. In this respect it is important to note that the same study found that a high level of the mH2A1.2 isoform was associated with a shorter metastasis-free disease survival in breast cancer.118

miRNA biogenesis

A DDX5/DDX17 function that is gaining considerable interest is their role in miRNA biogenesis. Several studies have demonstrated that DDX5 and DDX17 are components of the Drosha micro-processor complex and are important for the processing of certain subsets of miRNAs. Perhaps the most clear-cut evidence for the role of DDX5/DDX17 in miRNA biogenesis comes from a study that showed that transgenic mice in which either DDX5 or DDX17 had been knocked out showed defects in the processing of certain, but not all, miRNAs; interestingly DDX5 and DDX5 knockouts were associated with embryonic and post-natal lethality respectively, with the double knockout resulting in earlier embryonic lethality.79 Expression of certain miRNAs was reduced in both DDX5−/− and DDX17−/− embryos while for one miRNA tested (miR-214) expression was reduced in DDX17−/− but not DDX5−/− embryos. Taken together, these findings support the idea that DDX5 and DDX17 share some functions but also have other non-redundant functions in the cell. Interestingly, DDX5 was also shown to unwind the Let-7 miRNA precursor duplex and to be important for Let-7-mediated silencing,119 providing another mechanism by which it can modulate miRNA biogenesis.

It is established that deregulated miRNA expression can contribute to cancer development and progression (reviewed in ref. 120). Interestingly, DDX5 and DDX17 have been reported to modulate the association of p53,121 ERα 122, TGF-β and bone morphogenic protein (BMP)-specific SMAD signal transducers123 to the Drosha complex, and to regulate the processing of subsets of miRNAs that play key roles in cancer progression. However, DDX5 and DDX17 have been implicated in promoting the processing of both oncogenic and tumor/growth-suppressor miRNAs: DDX5 was found to promote processing of the oncogenic miR-21 via TGF-β/BMP123 but also of several growth suppressing miRNAs that are modulated by p53 (including miR16–1, miR-143 and miR-145).121 On the other hand, in a different context, DDX5 and DDX17 appear to enhance ERα-mediated inhibition of miR16–1, miR-143 and miR-145 processing.122 Such apparently opposing roles highlight the context dependence of DDX5/DDX17 function; e.g., expression of p53 and/or ERα. Furthermore, in basal breast cancer cells, DDX5 was found to regulate expression of several miRNAs including miR-21, miR-182 and Let-7 family miRNAs124; interestingly, siRNA knockdown of DDX5 or miR-182 inhibition resulted in actin cytoskeleton reorganization suggesting a role for DDX5 in cellular invasion through miR-182 regulation.

Ribosome biogenesis: rRNA processing

An additional finding from the DDX5/DDX17 knockout mouse study that described a role in Drosha miRNA processing was that DDX17−/− mouse embryo fibroblasts exhibited a reduced cell proliferation and an increase in cell death79; moreover in both DDX5−/− and DDX17−/− embryos there was a reduction of 5.8S rRNA expression, suggesting additional roles for DDX5 and DDX17 in rRNA processing. These findings are consistent with results obtained from DDX5/DDX17 siRNA knockdown from HeLa cells94 and from DDX5 siRNA knockdown in Mouse Embryo Fibroblasts study.95 The latter study also showed that DDX5 enhanced both the transcription of the 47S pre-rRNA and processing, underscoring the multi-functional roles of DDX5/DDX17. Furthermore, given that aberrant ribosome biogenesis can have profound effects on metabolism and cancer development/progression, these findings could have important implications for the potential role of DDX5 and DDX17 in cancer.

DEAD box RNA helicases in cancer: Is it all context?

It is clear that DEAD box RNA helicases generally function as components of multi-protein complexes and that they do not act by simply unwinding RNA duplexes; indeed several have been shown to act as RNPases or to exhibit RNA annealing activity (reviewed in ref. 125). Furthermore, while DEAD box proteins clearly have ATP-dependent functions through their shared helicase core domain, they have additional ATP-independent roles that are presumably conferred through their interaction with protein partners via their non-conserved N- and C-terminal domains. This would allow DEAD box proteins to integrate different processes in RNA metabolism: for example they could couple transcription, RNA processing and RNA export through ordered and regulated interactions with different protein partners. Such multi-functionality would, however, be highly context-dependent and would depend on the availability of interacting partners.

There are several key issues that need to be addressed regarding the precise mechanism(s) of action of DEAD box proteins. For example, it is established that several of these proteins act as transcriptional co-regulators, modulating transcriptional initiation through their interactions with components of the transcription machinery at responsive promoters, although there is no evidence that they directly bind chromatin. However, it is unclear what determines whether they act as co-activators or co-repressors. Furthermore, the mechanisms by which they might couple transcriptional initiation to post-transcriptional processes remain to be elucidated: it is not known whether these proteins bind the nascent RNAs and then act as ‘adaptor’ molecules to recruit splicing or export factors to facilitate these processes. In this respect it would also be interesting to examine the importance of helicase activity in these events since it appears to be required for some but not all the reported functions. Similarly, the precise roles of DEAD box proteins in other processes, e.g., regulation of translation or miRNA biogenesis, are not fully understood and much of the evidence relies on their association with the respective protein complexes and the effects of overexpression or siRNA knockdown studies.

As discussed above, several DEAD box proteins are aberrantly expressed in cancer tissues as compared with matched normal tissue and in some cases (e.g., DDX5) they also appear to be differentially post-translationally modified, supporting the idea that alterations in protein levels or modification can impact on their role in cell proliferation and/or transformation. However, the precise role of DEAD box proteins will clearly also depend on their interacting partners, the expression and/or function of which may also be independently altered during cancer development. In this respect, changes in post-translational modification of DEAD box proteins will also influence their interactions with partners and their impact on different cellular processes, adding further complexity. Thus a given DEAD box protein could have a growth promoting or pro-proliferation role in some contexts and a growth- or tumor-suppressing role in others. This context dependence would clearly have profound implications for the consideration of DEAD box proteins as possible biomarkers or therapeutic targets in cancer. Therefore, before choosing to therapeutically target a specific DEAD box protein it will be important to consider the type, or indeed subtype, of cancer and the expression of other interacting partners, since the therapy could have opposing effects in different contexts. There is clearly much work to be done in this area.

Acknowledgments

Work in the Fuller-Pace laboratory is supported by grants from Cancer Research UK and The Breast Cancer Campaign.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Footnotes

References

  • 1.Abdelhaleem M. Do human RNA helicases have a role in cancer? Biochim Biophys Acta. 2004;1704:37–46. doi: 10.1016/j.bbcan.2004.05.001. [DOI] [PubMed] [Google Scholar]
  • 2.Godbout R, Squire J. Amplification of a DEAD box protein gene in retinoblastoma cell lines. Proc Natl Acad Sci U S A. 1993;90:7578–82. doi: 10.1073/pnas.90.16.7578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.George RE, Kenyon RM, McGuckin AG, Malcolm AJ, Pearson AD, Lunec J, United Kingdom Children’s Cancer Study Group Investigation of co-amplification of the candidate genes ornithine decarboxylase, ribonucleotide reductase, syndecan-1 and a DEAD box gene, DDX1, with N-myc in neuroblastoma. Oncogene. 1996;12:1583–7. [PubMed] [Google Scholar]
  • 4.Manohar CF, Salwen HR, Brodeur GM, Cohn SL. Co-amplification and concomitant high levels of expression of a DEAD box gene with MYCN in human neuroblastoma. Genes Chromosomes Cancer. 1995;14:196–203. doi: 10.1002/gcc.2870140307. [DOI] [PubMed] [Google Scholar]
  • 5.Squire JA, Thorner PS, Weitzman S, Maggi JD, Dirks P, Doyle J, et al. Co-amplification of MYCN and a DEAD box gene (DDX1) in primary neuroblastoma. Oncogene. 1995;10:1417–22. [PubMed] [Google Scholar]
  • 6.George RE, Kenyon R, McGuckin AG, Kohl N, Kogner P, Christiansen H, et al. Analysis of candidate gene co-amplification with MYCN in neuroblastoma. Eur J Cancer. 1997;33:2037–42. doi: 10.1016/S0959-8049(97)00206-2. [DOI] [PubMed] [Google Scholar]
  • 7.Germain DR, Graham K, Glubrecht DD, Hugh JC, Mackey JR, Godbout R. DEAD box 1: a novel and independent prognostic marker for early recurrence in breast cancer. Breast Cancer Res Treat. 2011;127:53–63. doi: 10.1007/s10549-010-0943-7. [DOI] [PubMed] [Google Scholar]
  • 8.Weber A, Imisch P, Bergmann E, Christiansen H. Coamplification of DDX1 correlates with an improved survival probability in children with MYCN-amplified human neuroblastoma. J Clin Oncol. 2004;22:2681–90. doi: 10.1200/JCO.2004.07.192. [DOI] [PubMed] [Google Scholar]
  • 9.Taunk NK, Goyal S, Wu H, Moran MS, Chen S, Haffty BG. DEAD box 1 (DDX1) expression predicts for local control and overall survival in early stage, node-negative breast cancer. Cancer. 2012;118:888–98. doi: 10.1002/cncr.26352. [DOI] [PubMed] [Google Scholar]
  • 10.De Preter K, Speleman F, Combaret V, Lunec J, Board J, Pearson A, et al. No evidence for correlation of DDX1 gene amplification with improved survival probability in patients with MYCN-amplified neuroblastomas. J Clin Oncol. 2005;23:3167–8, author reply 3168-70. doi: 10.1200/JCO.2005.05.346. [DOI] [PubMed] [Google Scholar]
  • 11.De Preter K, Speleman F, Combaret V, Lunec J, Laureys G, Eussen BH, et al. Quantification of MYCN, DDX1, and NAG gene copy number in neuroblastoma using a real-time quantitative PCR assay. Mod Pathol. 2002;15:159–66. doi: 10.1038/modpathol.3880508. [DOI] [PubMed] [Google Scholar]
  • 12.Kaneko S, Ohira M, Nakamura Y, Isogai E, Nakagawara A, Kaneko M. Relationship of DDX1 and NAG gene amplification/overexpression to the prognosis of patients with MYCN-amplified neuroblastoma. J Cancer Res Clin Oncol. 2007;133:185–92. doi: 10.1007/s00432-006-0156-y. [DOI] [PubMed] [Google Scholar]
  • 13.de Souza DR, Sanabani SS, de Souza AC, Filho Odone V, Epelman S, Bendit I. Prognostic impact of MYCN, DDX1, TrkA, and TrkC gene transcripts expression in neuroblastoma. Pediatr Blood Cancer. 2011;56:749–56. doi: 10.1002/pbc.22823. [DOI] [PubMed] [Google Scholar]
  • 14.Godbout R, Li L, Liu RZ, Roy K. Role of DEAD box 1 in retinoblastoma and neuroblastoma. Future Oncol. 2007;3:575–87. doi: 10.2217/14796694.3.5.575. [DOI] [PubMed] [Google Scholar]
  • 15.Kanai Y, Dohmae N, Hirokawa N. Kinesin transports RNA: isolation and characterization of an RNA-transporting granule. Neuron. 2004;43:513–25. doi: 10.1016/j.neuron.2004.07.022. [DOI] [PubMed] [Google Scholar]
  • 16.Bléoo S, Sun X, Hendzel MJ, Rowe JM, Packer M, Godbout R. Association of human DEAD box protein DDX1 with a cleavage stimulation factor involved in 3′-end processing of pre-MRNA. Mol Biol Cell. 2001;12:3046–59. doi: 10.1091/mbc.12.10.3046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Li L, Monckton EA, Godbout R. A role for DEAD box 1 at DNA double-strand breaks. Mol Cell Biol. 2008;28:6413–25. doi: 10.1128/MCB.01053-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Buszczak M, Spradling AC. The Drosophila P68 RNA helicase regulates transcriptional deactivation by promoting RNA release from chromatin. Genes Dev. 2006;20:977–89. doi: 10.1101/gad.1396306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Konakahara S, Suzuki Y, Kawakami T, Saitou M, Kajikawa M, Masuho Y, et al. A neuronal transmembrane protein LRFN4 complexes with 14-3-3s and NCK1 to induce morphological change in monocytic cells via Rac1-mediated actin cytoskeleton reorganization. FEBS Lett. 2012;586:2251–9. doi: 10.1016/j.febslet.2012.05.053. [DOI] [PubMed] [Google Scholar]
  • 20.Meek SE, Lane WS, Piwnica-Worms H. Comprehensive proteomic analysis of interphase and mitotic 14-3-3-binding proteins. J Biol Chem. 2004;279:32046–54. doi: 10.1074/jbc.M403044200. [DOI] [PubMed] [Google Scholar]
  • 21.Pozuelo Rubio M, Geraghty KM, Wong BH, Wood NT, Campbell DG, Morrice N, et al. 14-3-3-affinity purification of over 200 human phosphoproteins reveals new links to regulation of cellular metabolism, proliferation and trafficking. Biochem J. 2004;379:395–408. doi: 10.1042/BJ20031797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Tanaka K, Okamoto S, Ishikawa Y, Tamura H, Hara T. DDX1 is required for testicular tumorigenesis, partially through the transcriptional activation of 12p stem cell genes. Oncogene. 2009;28:2142–51. doi: 10.1038/onc.2009.89. [DOI] [PubMed] [Google Scholar]
  • 23.Schröder M. Human DEAD-box protein 3 has multiple functions in gene regulation and cell cycle control and is a prime target for viral manipulation. Biochem Pharmacol. 2010;79:297–306. doi: 10.1016/j.bcp.2009.08.032. [DOI] [PubMed] [Google Scholar]
  • 24.Garbelli A, Radi M, Falchi F, Beermann S, Zanoli S, Manetti F, et al. Targeting the human DEAD-box polypeptide 3 (DDX3) RNA helicase as a novel strategy to inhibit viral replication. Curr Med Chem. 2011;18:3015–27. doi: 10.2174/092986711796391688. [DOI] [PubMed] [Google Scholar]
  • 25.Tarn WY, Chang TH. The current understanding of Ded1p/DDX3 homologs from yeast to human. RNA Biol. 2009;6:17–20. doi: 10.4161/rna.6.1.7440. [DOI] [PubMed] [Google Scholar]
  • 26.Stevens SW, Ryan DE, Ge HY, Moore RE, Young MK, Lee TD, et al. Composition and functional characterization of the yeast spliceosomal penta-snRNP. Mol Cell. 2002;9:31–44. doi: 10.1016/S1097-2765(02)00436-7. [DOI] [PubMed] [Google Scholar]
  • 27.Zhou Z, Licklider LJ, Gygi SP, Reed R. Comprehensive proteomic analysis of the human spliceosome. Nature. 2002;419:182–5. doi: 10.1038/nature01031. [DOI] [PubMed] [Google Scholar]
  • 28.Merz C, Urlaub H, Will CL, Lührmann R. Protein composition of human mRNPs spliced in vitro and differential requirements for mRNP protein recruitment. RNA. 2007;13:116–28. doi: 10.1261/rna.336807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yedavalli VS, Neuveut C, Chi YH, Kleiman L, Jeang KT. Requirement of DDX3 DEAD box RNA helicase for HIV-1 Rev-RRE export function. Cell. 2004;119:381–92. doi: 10.1016/j.cell.2004.09.029. [DOI] [PubMed] [Google Scholar]
  • 30.Lai MC, Lee YH, Tarn WY. The DEAD-box RNA helicase DDX3 associates with export messenger ribonucleoproteins as well as tip-associated protein and participates in translational control. Mol Biol Cell. 2008;19:3847–58. doi: 10.1091/mbc.E07-12-1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Choi YJ, Lee SG. The DEAD-box RNA helicase DDX3 interacts with DDX5, co-localizes with it in the cytoplasm during the G2/M phase of the cycle, and affects its shuttling during mRNP export. J Cell Biochem. 2012;113:985–96. doi: 10.1002/jcb.23428. [DOI] [PubMed] [Google Scholar]
  • 32.Chuang RY, Weaver PL, Liu Z, Chang TH. Requirement of the DEAD-Box protein ded1p for messenger RNA translation. Science. 1997;275:1468–71. doi: 10.1126/science.275.5305.1468. [DOI] [PubMed] [Google Scholar]
  • 33.de la Cruz J, Iost I, Kressler D, Linder P. The p20 and Ded1 proteins have antagonistic roles in eIF4E-dependent translation in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 1997;94:5201–6. doi: 10.1073/pnas.94.10.5201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shih JW, Tsai TY, Chao CH, Wu Lee YH. Candidate tumor suppressor DDX3 RNA helicase specifically represses cap-dependent translation by acting as an eIF4E inhibitory protein. Oncogene. 2008;27:700–14. doi: 10.1038/sj.onc.1210687. [DOI] [PubMed] [Google Scholar]
  • 35.Soto-Rifo R, Rubilar PS, Limousin T, de Breyne S, Décimo D, Ohlmann T. DEAD-box protein DDX3 associates with eIF4F to promote translation of selected mRNAs. EMBO J. 2012;31:3745–56. doi: 10.1038/emboj.2012.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Beckham C, Hilliker A, Cziko AM, Noueiry A, Ramaswami M, Parker R. The DEAD-box RNA helicase Ded1p affects and accumulates in Saccharomyces cerevisiae P-bodies. Mol Biol Cell. 2008;19:984–93. doi: 10.1091/mbc.E07-09-0954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hilliker A, Gao Z, Jankowsky E, Parker R. The DEAD-box protein Ded1 modulates translation by the formation and resolution of an eIF4F-mRNA complex. Mol Cell. 2011;43:962–72. doi: 10.1016/j.molcel.2011.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schröder M, Baran M, Bowie AG. Viral targeting of DEAD box protein 3 reveals its role in TBK1/IKKepsilon-mediated IRF activation. EMBO J. 2008;27:2147–57. doi: 10.1038/emboj.2008.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Soulat D, Bürckstümmer T, Westermayer S, Goncalves A, Bauch A, Stefanovic A, et al. The DEAD-box helicase DDX3X is a critical component of the TANK-binding kinase 1-dependent innate immune response. EMBO J. 2008;27:2135–46. doi: 10.1038/emboj.2008.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chao CH, Chen CM, Cheng PL, Shih JW, Tsou AP, Lee YH. DDX3, a DEAD box RNA helicase with tumor growth-suppressive property and transcriptional regulation activity of the p21waf1/cip1 promoter, is a candidate tumor suppressor. Cancer Res. 2006;66:6579–88. doi: 10.1158/0008-5472.CAN-05-2415. [DOI] [PubMed] [Google Scholar]
  • 41.Sun M, Song L, Zhou T, Gillespie GY, Jope RS. The role of DDX3 in regulating Snail. Biochim Biophys Acta. 2011;1813:438–47. doi: 10.1016/j.bbamcr.2011.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Botlagunta M, Vesuna F, Mironchik Y, Raman A, Lisok A, Winnard P, Jr., et al. Oncogenic role of DDX3 in breast cancer biogenesis. Oncogene. 2008;27:3912–22. doi: 10.1038/onc.2008.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Huang JS, Chao CC, Su TL, Yeh SH, Chen DS, Chen CT, et al. Diverse cellular transformation capability of overexpressed genes in human hepatocellular carcinoma. Biochem Biophys Res Commun. 2004;315:950–8. doi: 10.1016/j.bbrc.2004.01.151. [DOI] [PubMed] [Google Scholar]
  • 44.Lai MC, Chang WC, Shieh SY, Tarn WY. DDX3 regulates cell growth through translational control of cyclin E1. Mol Cell Biol. 2010;30:5444–53. doi: 10.1128/MCB.00560-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fukumura J, Noguchi E, Sekiguchi T, Nishimoto T. A temperature-sensitive mutant of the mammalian RNA helicase, DEAD-BOX X isoform, DBX, defective in the transition from G1 to S phase. J Biochem. 2003;134:71–82. doi: 10.1093/jb/mvg126. [DOI] [PubMed] [Google Scholar]
  • 46.Li Y, Wang H, Wang Z, Makhija S, Buchsbaum D, LoBuglio A, et al. Inducible resistance of tumor cells to tumor necrosis factor-related apoptosis-inducing ligand receptor 2-mediated apoptosis by generation of a blockade at the death domain function. Cancer Res. 2006;66:8520–8. doi: 10.1158/0008-5472.CAN-05-4364. [DOI] [PubMed] [Google Scholar]
  • 47.Sun M, Song L, Li Y, Zhou T, Jope RS. Identification of an antiapoptotic protein complex at death receptors. Cell Death Differ. 2008;15:1887–900. doi: 10.1038/cdd.2008.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Oliver PG, LoBuglio AF, Zhou T, Forero A, Kim H, Zinn KR, et al. Effect of anti-DR5 and chemotherapy on basal-like breast cancer. Breast Cancer Res Treat. 2012;133:417–26. doi: 10.1007/s10549-011-1755-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chang PC, Chi CW, Chau GY, Li FY, Tsai YH, Wu JC, et al. DDX3, a DEAD box RNA helicase, is deregulated in hepatitis virus-associated hepatocellular carcinoma and is involved in cell growth control. Oncogene. 2006;25:1991–2003. doi: 10.1038/sj.onc.1209239. [DOI] [PubMed] [Google Scholar]
  • 50.Wu DW, Liu WS, Wang J, Chen CY, Cheng YW, Lee H. Reduced p21(WAF1/CIP1) via alteration of p53-DDX3 pathway is associated with poor relapse-free survival in early-stage human papillomavirus-associated lung cancer. Clin Cancer Res. 2011;17:1895–905. doi: 10.1158/1078-0432.CCR-10-2316. [DOI] [PubMed] [Google Scholar]
  • 51.Lu D, Yunis JJ. Cloning, expression and localization of an RNA helicase gene from a human lymphoid cell line with chromosomal breakpoint 11q23.3. Nucleic Acids Res. 1992;20:1967–72. doi: 10.1093/nar/20.8.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Akao Y, Seto M, Yamamoto K, Iida S, Nakazawa S, Inazawa J, et al. The RCK gene associated with t(11;14) translocation is distinct from the MLL/ALL-1 gene with t(4;11) and t(11;19) translocations. Cancer Res. 1992;52:6083–7. [PubMed] [Google Scholar]
  • 53.Akao Y, Marukawa O, Morikawa H, Nakao K, Kamei M, Hachiya T, et al. The rck/p54 candidate proto-oncogene product is a 54-kilodalton D-E-A-D box protein differentially expressed in human and mouse tissues. Cancer Res. 1995;55:3444–9. [PubMed] [Google Scholar]
  • 54.Nakagawa Y, Morikawa H, Hirata I, Shiozaki M, Matsumoto A, Maemura K, et al. Overexpression of rck/p54, a DEAD box protein, in human colorectal tumours. Br J Cancer. 1999;80:914–7. doi: 10.1038/sj.bjc.6690441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hashimoto K, Nakagawa Y, Morikawa H, Niki M, Egashira Y, Hirata I, et al. Co-overexpression of DEAD box protein rck/p54 and c-myc protein in human colorectal adenomas and the relevance of their expression in cultured cell lines. Carcinogenesis. 2001;22:1965–70. doi: 10.1093/carcin/22.12.1965. [DOI] [PubMed] [Google Scholar]
  • 56.Miyaji K, Nakagawa Y, Matsumoto K, Yoshida H, Morikawa H, Hongou Y, et al. Overexpression of a DEAD box/RNA helicase protein, rck/p54, in human hepatocytes from patients with hepatitis C virus-related chronic hepatitis and its implication in hepatocellular carcinogenesis. J Viral Hepat. 2003;10:241–8. doi: 10.1046/j.1365-2893.2003.00447.x. [DOI] [PubMed] [Google Scholar]
  • 57.de Valoir T, Tucker MA, Belikoff EJ, Camp LA, Bolduc C, Beckingham K. A second maternally expressed Drosophila gene encodes a putative RNA helicase of the “DEAD box” family. Proc Natl Acad Sci U S A. 1991;88:2113–7. doi: 10.1073/pnas.88.6.2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nakamura A, Amikura R, Hanyu K, Kobayashi S. Me31B silences translation of oocyte-localizing RNAs through the formation of cytoplasmic RNP complex during Drosophila oogenesis. Development. 2001;128:3233–42. doi: 10.1242/dev.128.17.3233. [DOI] [PubMed] [Google Scholar]
  • 59.Maekawa H, Nakagawa T, Uno Y, Kitamura K, Shimoda C. The ste13+ gene encoding a putative RNA helicase is essential for nitrogen starvation-induced G1 arrest and initiation of sexual development in the fission yeast Schizosaccharomyces pombe. Mol Gen Genet. 1994;244:456–64. doi: 10.1007/BF00583896. [DOI] [PubMed] [Google Scholar]
  • 60.Ladomery M, Wade E, Sommerville J. Xp54, the Xenopus homologue of human RNA helicase p54, is an integral component of stored mRNP particles in oocytes. Nucleic Acids Res. 1997;25:965–73. doi: 10.1093/nar/25.5.965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Navarro RE, Shim EY, Kohara Y, Singson A, Blackwell TK. cgh-1, a conserved predicted RNA helicase required for gametogenesis and protection from physiological germline apoptosis in C. elegans. Development. 2001;128:3221–32. doi: 10.1242/dev.128.17.3221. [DOI] [PubMed] [Google Scholar]
  • 62.Matsumoto K, Kwon OY, Kim H, Akao Y. Expression of rck/p54, a DEAD-box RNA helicase, in gametogenesis and early embryogenesis of mice. Dev Dyn. 2005;233:1149–56. doi: 10.1002/dvdy.20429. [DOI] [PubMed] [Google Scholar]
  • 63.Weston A, Sommerville J. Xp54 and related (DDX6-like) RNA helicases: roles in messenger RNP assembly, translation regulation and RNA degradation. Nucleic Acids Res. 2006;34:3082–94. doi: 10.1093/nar/gkl409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Smillie DA, Sommerville J. RNA helicase p54 (DDX6) is a shuttling protein involved in nuclear assembly of stored mRNP particles. J Cell Sci. 2002;115:395–407. doi: 10.1242/jcs.115.2.395. [DOI] [PubMed] [Google Scholar]
  • 65.Minshall N, Thom G, Standart N. A conserved role of a DEAD box helicase in mRNA masking. RNA. 2001;7:1728–42. doi: 10.1017/S135583820101158X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Minshall N, Kress M, Weil D, Standart N. Role of p54 RNA helicase activity and its C-terminal domain in translational repression, P-body localization and assembly. Mol Biol Cell. 2009;20:2464–72. doi: 10.1091/mbc.E09-01-0035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chu CY, Rana TM. Translation repression in human cells by microRNA-induced gene silencing requires RCK/p54. PLoS Biol. 2006;4:e210. doi: 10.1371/journal.pbio.0040210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Coller JM, Tucker M, Sheth U, Valencia-Sanchez MA, Parker R. The DEAD box helicase, Dhh1p, functions in mRNA decapping and interacts with both the decapping and deadenylase complexes. RNA. 2001;7:1717–27. doi: 10.1017/S135583820101994X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Coller J, Parker R. General translational repression by activators of mRNA decapping. Cell. 2005;122:875–86. doi: 10.1016/j.cell.2005.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ernoult-Lange M, Baconnais S, Harper M, Minshall N, Souquere S, Boudier T, et al. Multiple binding of repressed mRNAs by the P-body protein Rck/p54. RNA. 2012;18:1702–15. doi: 10.1261/rna.034314.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chang LC, Lee FJ. The RNA helicase Dhh1p cooperates with Rbp1p to promote porin mRNA decay via its non-conserved C-terminal domain. Nucleic Acids Res. 2012;40:1331–44. doi: 10.1093/nar/gkr803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bergkessel M, Reese JC. An essential role for the Saccharomyces cerevisiae DEAD-box helicase DHH1 in G1/S DNA-damage checkpoint recovery. Genetics. 2004;167:21–33. doi: 10.1534/genetics.167.1.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Akao Y, Matsumoto K, Ohguchi K, Nakagawa Y, Yoshida H. Human DEAD-box/RNA unwindase rck/p54 contributes to maintenance of cell growth by affecting cell cycle in cultured cells. Int J Oncol. 2006;29:41–8. [PubMed] [Google Scholar]
  • 74.Lin F, Wang R, Shen JJ, Wang X, Gao P, Dong K, et al. Knockdown of RCK/p54 expression by RNAi inhibits proliferation of human colorectal cancer cells in vitro and in vivo. Cancer Biol Ther. 2008;7:1669–76. doi: 10.4161/cbt.7.10.6660. [DOI] [PubMed] [Google Scholar]
  • 75.Akao Y, Mizoguchi H, Ohishi N, Yagi K. Growth inhibition by overexpression of human DEAD box protein rck/p54 in cells of a guinea pig cell line. FEBS Lett. 1998;429:279–83. doi: 10.1016/S0014-5793(98)00605-X. [DOI] [PubMed] [Google Scholar]
  • 76.Ford MJ, Anton IA, Lane DP. Nuclear protein with sequence homology to translation initiation factor eIF-4A. Nature. 1988;332:736–8. doi: 10.1038/332736a0. [DOI] [PubMed] [Google Scholar]
  • 77.Lamm GM, Nicol SM, Fuller-Pace FV, Lamond AI. p72: a human nuclear DEAD box protein highly related to p68. Nucleic Acids Res. 1996;24:3739–47. doi: 10.1093/nar/24.19.3739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fuller-Pace FV, Moore HC. RNA helicases p68 and p72: multifunctional proteins with important implications for cancer development. Future Oncol. 2011;7:239–51. doi: 10.2217/fon.11.1. [DOI] [PubMed] [Google Scholar]
  • 79.Fukuda T, Yamagata K, Fujiyama S, Matsumoto T, Koshida I, Yoshimura K, et al. DEAD-box RNA helicase subunits of the Drosha complex are required for processing of rRNA and a subset of microRNAs. Nat Cell Biol. 2007;9:604–11. doi: 10.1038/ncb1577. [DOI] [PubMed] [Google Scholar]
  • 80.Stevenson RJ, Hamilton SJ, MacCallum DE, Hall PA, Fuller-Pace FV. Expression of the ‘dead box’ RNA helicase p68 is developmentally and growth regulated and correlates with organ differentiation/maturation in the fetus. J Pathol. 1998;184:351–9. doi: 10.1002/(SICI)1096-9896(199804)184:4<351::AID-PATH1235>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 81.Causevic M, Hislop RG, Kernohan NM, Carey FA, Kay RA, Steele RJ, et al. Overexpression and poly-ubiquitylation of the DEAD-box RNA helicase p68 in colorectal tumours. Oncogene. 2001;20:7734–43. doi: 10.1038/sj.onc.1204976. [DOI] [PubMed] [Google Scholar]
  • 82.Yang L, Lin C, Liu ZR. Phosphorylations of DEAD box p68 RNA helicase are associated with cancer development and cell proliferation. Mol Cancer Res. 2005;3:355–63. doi: 10.1158/1541-7786.MCR-05-0022. [DOI] [PubMed] [Google Scholar]
  • 83.Shin S, Rossow KL, Grande JP, Janknecht R. Involvement of RNA helicases p68 and p72 in colon cancer. Cancer Res. 2007;67:7572–8. doi: 10.1158/0008-5472.CAN-06-4652. [DOI] [PubMed] [Google Scholar]
  • 84.Clark EL, Coulson A, Dalgliesh C, Rajan P, Nicol SM, Fleming S, et al. The RNA helicase p68 is a novel androgen receptor coactivator involved in splicing and is overexpressed in prostate cancer. Cancer Res. 2008;68:7938–46. doi: 10.1158/0008-5472.CAN-08-0932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wortham NC, Ahamed E, Nicol SM, Thomas RS, Periyasamy M, Jiang J, et al. The DEAD-box protein p72 regulates ERalpha-/oestrogen-dependent transcription and cell growth, and is associated with improved survival in ERalpha-positive breast cancer. Oncogene. 2009;28:4053–64. doi: 10.1038/onc.2009.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wang SJ, Zhang C, You Y, Shi CM. Overexpression of RNA helicase p68 protein in cutaneous squamous cell carcinoma. Clin Exp Dermatol. 2012;37:882–8. doi: 10.1111/j.1365-2230.2012.04365.x. [DOI] [PubMed] [Google Scholar]
  • 87.Wang R, Jiao Z, Li R, Yue H, Chen L. p68 RNA helicase promotes glioma cell proliferation in vitro and in vivo via direct regulation of NF-κB transcription factor p50. Neuro Oncol. 2012;14:1116–24. doi: 10.1093/neuonc/nos131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yang L, Lin C, Liu ZR. P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing Axin from beta-catenin. Cell. 2006;127:139–55. doi: 10.1016/j.cell.2006.08.036. [DOI] [PubMed] [Google Scholar]
  • 89.Iqbal S, Zhang S, Driss A, Liu ZR, Kim HR, Wang Y, et al. PDGF upregulates Mcl-1 through activation of β-catenin and HIF-1α-dependent signaling in human prostate cancer cells. PLoS One. 2012;7:e30764. doi: 10.1371/journal.pone.0030764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Stucke VM, Gorses D, Hofmann F. DEAD-box RNA helicase p68 is not required for nuclear translocation of beta-catenin in colon cancer cells. Cell Cycle. 2008;7:830–2. doi: 10.4161/cc.7.6.5614. [DOI] [PubMed] [Google Scholar]
  • 91.Yang L, Lin C, Zhao S, Wang H, Liu ZR. Phosphorylation of p68 RNA helicase plays a role in platelet-derived growth factor-induced cell proliferation by up-regulating cyclin D1 and c-Myc expression. J Biol Chem. 2007;282:16811–9. doi: 10.1074/jbc.M610488200. [DOI] [PubMed] [Google Scholar]
  • 92.Carter CL, Lin C, Liu CY, Yang L, Liu ZR. Phosphorylated p68 RNA helicase activates Snail1 transcription by promoting HDAC1 dissociation from the Snail1 promoter. Oncogene. 2010;29:5427–36. doi: 10.1038/onc.2010.276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yang L, Lin C, Sun SY, Zhao S, Liu ZR. A double tyrosine phosphorylation of P68 RNA helicase confers resistance to TRAIL-induced apoptosis. Oncogene. 2007;26:6082–92. doi: 10.1038/sj.onc.1210427. [DOI] [PubMed] [Google Scholar]
  • 94.Jalal C, Uhlmann-Schiffler H, Stahl H. Redundant role of DEAD box proteins p68 (Ddx5) and p72/p82 (Ddx17) in ribosome biogenesis and cell proliferation. Nucleic Acids Res. 2007;35:3590–601. doi: 10.1093/nar/gkm058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Saporita AJ, Chang HC, Winkeler CL, Apicelli AJ, Kladney RD, Wang J, et al. RNA helicase DDX5 is a p53-independent target of ARF that participates in ribosome biogenesis. Cancer Res. 2011;71:6708–17. doi: 10.1158/0008-5472.CAN-11-1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mazurek A, Luo W, Krasnitz A, Hicks J, Powers RS, Stillman B. DDX5 regulates DNA replication and is required for cell proliferation in a subset of breast cancer cells. Cancer Discov. 2012;2:812–25. doi: 10.1158/2159-8290.CD-12-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Endoh H, Maruyama K, Masuhiro Y, Kobayashi Y, Goto M, Tai H, et al. Purification and identification of p68 RNA helicase acting as a transcriptional coactivator specific for the activation function 1 of human estrogen receptor alpha. Mol Cell Biol. 1999;19:5363–72. doi: 10.1128/mcb.19.8.5363. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 98.Jensen ED, Niu L, Caretti G, Nicol SM, Teplyuk N, Stein GS, et al. p68 (Ddx5) interacts with Runx2 and regulates osteoblast differentiation. J Cell Biochem. 2008;103:1438–51. doi: 10.1002/jcb.21526. [DOI] [PubMed] [Google Scholar]
  • 99.Caretti G, Schiltz RL, Dilworth FJ, Di Padova M, Zhao P, Ogryzko V, et al. The RNA helicases p68/p72 and the noncoding RNA SRA are coregulators of MyoD and skeletal muscle differentiation. Dev Cell. 2006;11:547–60. doi: 10.1016/j.devcel.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 100.Bates GJ, Nicol SM, Wilson BJ, Jacobs AM, Bourdon JC, Wardrop J, et al. The DEAD box protein p68: a novel transcriptional coactivator of the p53 tumour suppressor. EMBO J. 2005;24:543–53. doi: 10.1038/sj.emboj.7600550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nicol SM, Bray SE, Derek Black H, Lorimore SA, Wright EG, Lane DP, et al. The RNA helicase p68 (DDX5) is selectively required for the induction of p53-dependent p21 expression and cell-cycle arrest after DNA damage. Oncogene. 2012 doi: 10.1038/onc.2012.426. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wagner M, Rid R, Maier CJ, Maier RH, Laimer M, Hintner H, et al. DDX5 is a multifunctional co-activator of steroid hormone receptors. Mol Cell Endocrinol. 2012;361:80–91. doi: 10.1016/j.mce.2012.03.014. [DOI] [PubMed] [Google Scholar]
  • 103.Jacobs AM, Nicol SM, Hislop RG, Jaffray EG, Hay RT, Fuller-Pace FV. SUMO modification of the DEAD box protein p68 modulates its transcriptional activity and promotes its interaction with HDAC1. Oncogene. 2007;26:5866–76. doi: 10.1038/sj.onc.1210387. [DOI] [PubMed] [Google Scholar]
  • 104.Mooney SM, Grande JP, Salisbury JL, Janknecht R. Sumoylation of p68 and p72 RNA helicases affects protein stability and transactivation potential. Biochemistry. 2010;49:1–10. doi: 10.1021/bi901263m. [DOI] [PubMed] [Google Scholar]
  • 105.Mooney SM, Goel A, D’Assoro AB, Salisbury JL, Janknecht R. Pleiotropic effects of p300-mediated acetylation on p68 and p72 RNA helicase. J Biol Chem. 2010;285:30443–52. doi: 10.1074/jbc.M110.143792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wilson BJ, Bates GJ, Nicol SM, Gregory DJ, Perkins ND, Fuller-Pace FV. The p68 and p72 DEAD box RNA helicases interact with HDAC1 and repress transcription in a promoter-specific manner. BMC Mol Biol. 2004;5:11. doi: 10.1186/1471-2199-5-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Guo J, Hong F, Loke J, Yea S, Lim CL, Lee U, et al. A DDX5 S480A polymorphism is associated with increased transcription of fibrogenic genes in hepatic stellate cells. J Biol Chem. 2010;285:5428–37. doi: 10.1074/jbc.M109.035295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Germann S, Gratadou L, Zonta E, Dardenne E, Gaudineau B, Fougère M, et al. Dual role of the ddx5/ddx17 RNA helicases in the control of the pro-migratory NFAT5 transcription factor. Oncogene. 2012;31:4536–49. doi: 10.1038/onc.2011.618. [DOI] [PubMed] [Google Scholar]
  • 109.Yao H, Brick K, Evrard Y, Xiao T, Camerini-Otero RD, Felsenfeld G. Mediation of CTCF transcriptional insulation by DEAD-box RNA-binding protein p68 and steroid receptor RNA activator SRA. Genes Dev. 2010;24:2543–55. doi: 10.1101/gad.1967810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dutertre M, Gratadou L, Dardenne E, Germann S, Samaan S, Lidereau R, et al. Estrogen regulation and physiopathologic significance of alternative promoters in breast cancer. Cancer Res. 2010;70:3760–70. doi: 10.1158/0008-5472.CAN-09-3988. [DOI] [PubMed] [Google Scholar]
  • 111.Liu ZR, Sargueil B, Smith CW. Detection of a novel ATP-dependent cross-linked protein at the 5′ splice site-U1 small nuclear RNA duplex by methylene blue-mediated photo-cross-linking. Mol Cell Biol. 1998;18:6910–20. doi: 10.1128/mcb.18.12.6910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Liu ZR. p68 RNA helicase is an essential human splicing factor that acts at the U1 snRNA-5′ splice site duplex. Mol Cell Biol. 2002;22:5443–50. doi: 10.1128/MCB.22.15.5443-5450.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lee CG. RH70, a bidirectional RNA helicase, co-purifies with U1snRNP. J Biol Chem. 2002;277:39679–83. doi: 10.1074/jbc.C200337200. [DOI] [PubMed] [Google Scholar]
  • 114.Auboeuf D, Hönig A, Berget SM, O’Malley BW. Coordinate regulation of transcription and splicing by steroid receptor coregulators. Science. 2002;298:416–9. doi: 10.1126/science.1073734. [DOI] [PubMed] [Google Scholar]
  • 115.Hönig A, Auboeuf D, Parker MM, O’Malley BW, Berget SM. Regulation of alternative splicing by the ATP-dependent DEAD-box RNA helicase p72. Mol Cell Biol. 2002;22:5698–707. doi: 10.1128/MCB.22.16.5698-5707.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Guil S, Gattoni R, Carrascal M, Abián J, Stévenin J, Bach-Elias M. Roles of hnRNP A1, SR proteins, and p68 helicase in c-H-ras alternative splicing regulation. Mol Cell Biol. 2003;23:2927–41. doi: 10.1128/MCB.23.8.2927-2941.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Camats M, Guil S, Kokolo M, Bach-Elias M. P68 RNA helicase (DDX5) alters activity of cis- and trans-acting factors of the alternative splicing of H-Ras. PLoS One. 2008;3:e2926. doi: 10.1371/journal.pone.0002926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Dardenne E, Pierredon S, Driouch K, Gratadou L, Lacroix-Triki M, Espinoza MP, et al. Splicing switch of an epigenetic regulator by RNA helicases promotes tumor-cell invasiveness. Nat Struct Mol Biol. 2012;19:1139–46. doi: 10.1038/nsmb.2390. [DOI] [PubMed] [Google Scholar]
  • 119.Salzman DW, Shubert-Coleman J, Furneaux H. P68 RNA helicase unwinds the human let-7 microRNA precursor duplex and is required for let-7-directed silencing of gene expression. J Biol Chem. 2007;282:32773–9. doi: 10.1074/jbc.M705054200. [DOI] [PubMed] [Google Scholar]
  • 120.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009;10:704–14. doi: 10.1038/nrg2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Suzuki HI, Yamagata K, Sugimoto K, Iwamoto T, Kato S, Miyazono K. Modulation of microRNA processing by p53. Nature. 2009;460:529–33. doi: 10.1038/nature08199. [DOI] [PubMed] [Google Scholar]
  • 122.Yamagata K, Fujiyama S, Ito S, Ueda T, Murata T, Naitou M, et al. Maturation of microRNA is hormonally regulated by a nuclear receptor. Mol Cell. 2009;36:340–7. doi: 10.1016/j.molcel.2009.08.017. [DOI] [PubMed] [Google Scholar]
  • 123.Davis BN, Hilyard AC, Lagna G, Hata A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature. 2008;454:56–61. doi: 10.1038/nature07086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wang D, Huang J, Hu Z. RNA helicase DDX5 regulates microRNA expression and contributes to cytoskeletal reorganization in basal breast cancer cells. Mol Cell Proteomics. 2012;11:M111–, 011932. doi: 10.1074/mcp.M111.011932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Linder P, Jankowsky E. From unwinding to clamping - the DEAD box RNA helicase family. Nat Rev Mol Cell Biol. 2011;12:505–16. doi: 10.1038/nrm3154. [DOI] [PubMed] [Google Scholar]

Articles from RNA Biology are provided here courtesy of Taylor & Francis

RESOURCES