Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 28.
Published in final edited form as: Wiley Interdiscip Rev RNA. 2010 May 6;1(2):10.1002/wrna.4. doi: 10.1002/wrna.4

Post-transcriptional regulation of cancer traits by HuR

Kotb Abdelmohsen 1, Myriam Gorospe 1,*
PMCID: PMC3808850  NIHMSID: NIHMS508441  PMID: 21935886

Abstract

Cancer-related gene expression programs are strongly influenced by post-transcriptional mechanisms. The RNA-binding protein HuR is highly abundant in many cancers. Numerous HuR-regulated mRNAs encode proteins implicated in carcinogenesis. Here, we review the collections of HuR target mRNAs that encode proteins responsible for implementing five major cancer traits. By interacting with specific mRNA subsets, HuR enhances the levels of proteins that 1) promote cell proliferation, 2) increase cell survival, 3) elevate local angiogenesis, 4) help the cancer cell evade immune recognition, and 5) facilitate cancer cell invasion and metastasis. We propose that HuR exerts a tumorigenic function by enabling these cancer phenotypes. We discuss evidence that links HuR to several specific cancers and suggests its potential usefulness in cancer diagnosis, prognosis, and therapy.

INTRODUCTION

The expression of distinct collections of proteins allows cancer cells to develop, survive, proliferate, and colonize other tissues (1). Although some changes in the expression of cancer-related proteins arise through gene mutation, most cancer-specific protein expression patterns involve genes that are not mutated, but are expressed aberrantly (2). Besides modified transcription, protein production can change through altered post-transcriptional mechanisms such as mRNA splicing, transport, storage, translation, and degradation (35). These processes are governed by two types of RNA-binding factors, RNA-binding proteins (RBPs) and noncoding RNAs, particularly microRNAs (6, 7).

The impact of RBPs on the expression of cancer-associated genes is well recognized. Many oncoproteins, tumor suppressor proteins, and other cancer-related proteins are encoded by mRNAs whose half-lives and/or translation are tightly regulated. The sequences that confer differential turnover or translation typically reside in the 5′- and 3′-untranslated regions (UTRs) of the mRNA. Accordingly, several RBPs that associate with these regions and modulate mRNA turnover and translation (e.g., tristetraprolin, AUF1, and Sam68) have been found to influence cancer-associated protein expression (3, 4, 8, 9). Among the earliest RBPs that were identified as being associated with tumorigenesis (10) are the members of the Hu/elav (embryonic lethal abnormal vision) protein family, which comprises three primarily neuronal proteins (HuB, HuC, and HuD), and one ubiquitous protein, HuR [ELAVL1, also known as HuA (11, 12)].

HuR FUNCTION

HuR has three RNA recognition motifs (RRMs) through which it interacts with target mRNAs, preferentially those bearing U- or AU-rich sequences in their 5′UTRs and 3′UTRs (11, 13). The association of HuR and target transcripts is modulated following phosphorylation by protein kinase C (PKC), the mitogen-activated protein kinase (MAPK) p38, and the checkpoint kinase Chk2, which also modulates HuR levels by influencing its ubiquitination (1418). HuR is primarily nuclear and has been implicated in mRNA splicing (19), but its nuclear functions remain obscure. By contrast, HuR’s ability to stabilize and/or modulate the translation of many of its target mRNAs is closely linked to its translocation to the cytoplasm (11). The transport of HuR across the nuclear envelope involves a specific HuR domain (the HuR nucleocytoplasmic shuttling sequence or HNS) and several transport machinery components, including CRM1, transportins 1 and 2, and importin-1α (2023). HuR nucleocytoplasmic transport is also influenced by kinases [cyclin-dependent kinase (Cdk)1, AMP-activated protein kinase (AMPK), PKC, and p38] that phosphorylate HuR and HuR transport components (15, 16, 2427). These and other post-translational modifications (28, 29) of HuR that affect HuR abundance, subcellular localization, and mRNA binding are summarized in Fig. 1.

Figure 1. HuR protein and post-translational modification by cancer-related enzymes.

Figure 1

The HuR three RNA recognition motifs (RRMs) and hinge region (amino acids 186-244), containing the HuR nucleocytoplasmic shuttling (HNS) domain, are indicated. The specific residues (Amino Acid Positions, first column) implicated in different post-translational modifications (second column) and the cancer-related enzymes that carry out the modifications (third column) are listed. The consequences of modification at each residue are listed under ‘Impact on HuR Function’. n.d., not determined.

HuR-stabilized target mRNAs include those that encode p21, c-fos, vascular endothelial growth factor (VEGF), the MAPK phosphatase (MKP)-1, inducible nitric oxide synthase (iNOS), granulocyte/macrophage-colony-stimulating factor (GM-CSF), sirtuin 1, tumor necrosis factor (TNF)-α, Bcl-2, Mcl-1, COX-2, γ-glutamylcysteine synthetase heavy subunit (γ-GCSh), urokinase-type plasminogen activator (uPA) and its receptor (uPAR), p53, interleukin (IL)-3, and cyclins A2, B1, E, and D1 (14, 3041). The exact mechanisms by which HuR stabilizes these mRNAs are not fully understood, but HuR likely competes with other RBPs whose interaction with the same labile mRNAs might lead to their recruitment to cellular structures for mRNA degradation, such as the exosome and processing (P) bodies (4249).

HuR can repress the translation of many target mRNAs, like those encoding p27, the type-I insulin-like growth factor receptor (IGF-IR), Wnt5a, and c-Myc (41, 5053). Additionally, HuR can promote the translation of many other target transcripts, such as those that encode the hypoxia-inducible factor (HIF)-1α, p53, prothymosin α (ProTα), MKP-1, cytochrome c, heme oxygenase-1, and CAT-1 (5459). The molecular mechanisms whereby HuR modulates translation are also poorly characterized. In some cases, HuR was proposed to interfere with internal ribosome entry sites (IRESs) in the 5′UTRs of target mRNAs (50, 52); in other cases, its effects on translation were due to competition or cooperation with microRNAs (53, 56).

Through its influence on collections of expressed proteins, HuR has been implicated in many cellular processes, including differentiation, the cellular response to damaging stimuli, and the immune and inflammatory responses (reviewed in 11, 41). In addition, HuR is increasingly recognized as a pivotal factor in cancer-related gene expression. This function is based on HuR’s ability to promote the expression of proteins that enhance proliferation, inhibit apoptosis, increase angiogenesis, reduce immune recognition, and facilitate invasion and metastasis (Table 1). Accordingly, a direct role for HuR in tumorigenesis is beginning to emerge (11, 60, 61). In this review, we will first describe the subsets of HuR target transcripts that promote each of these traits of cancer cells. We will then describe the evidence linking HuR to specific malignancies and its growing recognition as a diagnostic, prognostic, and therapeutic target in human cancer.

Table 1. Specific regulation of HuR-target mRNAs encoding cancer proteins.

Listed are HuR target transcripts, the regions (3′UTR, 5′UTR) where the binding sites are located, and HuR’s influence on the stability or translation of each mRNA. The main cancer traits influenced by the encoded proteins are indicated under ‘Main Cancer Traits’.

Target mRNA Binding Site Influence on mRNA Main Cancer Traits Reference
Cyclin D1 3′UTR Stability ↑ Proliferation (37)
Cyclin E1 3′UTR Stability ↑ Proliferation (74,75)
Cyclin A2 3′UTR Stability ↑ Translation ↑ Proliferation (25,26,31)
Cyclin B1 3′UTR Stability ↑ Proliferation (26,31,66)
p27 5′UTR Translation ↓ Proliferation, Survival (50)
EGF 3′UTR ↑ (n.d.) Proliferation, Survival (83)
eIF4E 3′UTR Stability ↑ Proliferation, Survival (86)
ProTα 3′UTR Translation ↑ Proliferation, Survival (55)
Bcl-2 3′UTR Stability ↑ Translation ↑ Survival (39,91)
Mcl-1 n.d. Stability ↑ Survival (39)
SIRT1 3′UTR Stability ↑ Survival (14)
p21 3′UTR Stability ↑ Proliferation, Survival (17,32)
Mdm-2 n.d. Stability ↑ Survival (100)
c-Myc 3′UTR Translation ↓ Proliferation, Survival (53,103)
HIF-1α 5′UTR, 3′UTR Stability ↑ Angiogenesis, Survival (83,111)
VEGF 3′UTR Stability ↑ Translation ↑ Angiogenesis, Proliferation (30,117)
COX-2 3′UTR Stability ↑ Angiogenesis (120,121)
TSP1* 3′UTR Translation ↑ Angiogenesis (62)
TGF-β 3′UTR ↑ (n.d.) Immunity/inflammation (136)
Snail 3′UTR Stability ↑ Invasion (138)
MMP-9 3′UTR Stability ↑ Invasion (146, 147)
uPA 3′UTR Stability ↑ Invasion (36)
uPAR 3′UTR Stability ↑ Invasion (36)

BRCA1 3′UTR ↓ (n.d.) Tumor suppression (156)
Wnt5a 3′UTR Translation ↓ Oncogenesis (52)
c-fms 3′UTR Stability ↑ Oncogenesis (154)
ARHI 3′UTR Stability ↑ Tumor suppression (159)

Bottom, HuR target mRNAs encoding proteins with broad cancer-related functions. ↑, HuR stabilizes or increases translation; ↓, HuR represses translation;

*

the interaction of HuR with TSP1 mRNA declines in cancer. n.d., molecular mechanisms not determined.

HuR-REGULATED mRNAs ENCODING PROTEINS IMPLICATED IN CANCER TRAITS

As proposed by Hanahan and Weinberg, normal cells evolve into cancer cells through the acquisition of specific phenotypes, including enhanced abilities to proliferate, survive apoptotic stimuli, develop local angiogenesis, and colonize other tissues (1). Over the past decade, numerous HuR-regulated mRNAs have been identified that contribute directly to the acquisition of each of these cancer traits (11, 61, 62). In this section, we will review these hallmark features of cancer cells and identify the HuR target mRNAs through which HuR can elicit cancer-related phenotypes (Fig. 2).

Figure 2. HuR-target mRNAs implicated in establishing cancer traits.

Figure 2

The subsets of HuR target mRNAs involved in five major cancer-acquired phenotypes are listed. * denotes transcripts whose expression decreases in the presence of HuR, either because HuR represses their translation (c-Myc, p27) or because its association with HuR is reduced in cancer (TSP1).

Promotion of cell proliferation

Cancer cells must divide actively in order for the tumor mass to grow. This trait is usually accompanied by altered abundance of cell cycle regulators, leading to shortened cell division times and to expansion of the tumor population. By associating with the mRNAs that encode them, HuR increases the levels of many cell cycle regulators, particularly cyclins, which activate cdks during different cell cycle phases; cyclins and cdks have been directly implicated in cancer progression (63). Additionally, the process of cellular senescence, widely considered to be a tumor suppressive mechanism linked to the cessation of cell division (64, 65), is suppressed by HuR overexpression (66). HuR inhibits cellular replicative senescence at least in part by enhancing the expression of several cyclins (66).

Cyclin D1

This cyclin is an essential activator of Cdk4 and Cdk6, which participate in the progression through the G1 phase of the cell division cycle. Elevated Cyclin D1 expression has been reported in tumor cells, associated with a shortened G1 phase (67, 68). HuR associates with the 3′UTR of Cyclin D1 mRNA and stabilizes it, at least partly by competing with decay-promoting RBPs, as observed in human cervical carcinoma cells (37).

Cyclin E

An activator of Cdk2, Cyclin E is critical for progression through the G1/S transition. Cyclin E is highly expressed in several malignancies, including gastric, ovarian, breast and colorectal cancers, and plays an important role in oncogenic transformation (7073). HuR was recently found to bind to the Cyclin E 3′UTR and promoted Cyclin E expression in the breast cancer cells (74, 75). Downregulation of HuR significantly lowered Cyclin E mRNA stability and protein levels, while HuR overexpression elevated Cyclin E1 production, in turn accelerating cell proliferation (74, 75).

Cyclin A2

Also a Cdk2 cofactor, Cyclin A2 promotes progression through the S phase. Cyclin A2 levels increase in tumors, associated with elevated proliferation (76, 77). HuR was shown to interact with the Cyclin A2 3′UTR and stabilized the Cyclin A2 mRNA in a cell cycle-dependent manner in colon cancer cells (31). In cervical carcinoma cells, HuR phosphorylation at serine 202 (S202) by the G2 kinase Cdk1 caused the nuclear retention of HuR, leading to marked reductions in both Cyclin A2 mRNA stability and Cyclin A2 translation (25). Conversely, the nonphosphorylatable mutants HuR(S202A) and HuR(S242A) accumulated in the cytoplasm, enhancing Cyclin A2 expression and increasing cell proliferation (2527).

Cyclin B1

A major activator of Cdk1 and hence a key factor for progression through the G2 phase, cyclin B1 abundance is elevated in tumors (78, 79). HuR associated with the 3′UTR of the cyclin B1 mRNA and stabilized it during the S and G2 phases in colon cancer cells (31, 66). As described for Cyclin A2 mRNA, the cytoplasmic accumulation of the nonphosphorylatable mutant HuR(S242A) also led to the stabilization of Cyclin B1 mRNA (26).

p27

By inhibiting Cdk2, p27 prevents cell proliferation (80). Reductions in p27 levels are associated with cancer, both because low p27 levels enhance cell division and because p27 is strongly pro-apoptotic (81). HuR interacts with the 5′UTR of p27 mRNA and represses p27 translation (50); the precise mechanisms of repression is unclear, but HuR may disrupt the activity of an IRES in the p27 5′UTR (50).

Epidermal growth factor (EGF)

Besides acting on cell cycle regulatory factors, HuR can promote the expression of factors that promote proliferation. EGF is a key signaling molecule that stimulates cell growth and proliferation. It functions by interacting with the EGF receptor (EGFR), which triggers proliferative signaling pathways and promotes tumorigenesis (82). HuR associates with the EGF 3′UTR, although its exact influence upon EGF expression awaits further study (83).

Eukaryotic translation initiation factor (eIF4E)

Eukaryotic mRNAs possess a 7-methyl-guanosine (m7G) cap structure at their 5′ end. The protein eIF4E binds the m7G cap and brings mRNAs to the ribosome for translation (84). eIF4E is found elevated in human cancers, is considered to be oncogenic, since its overexpression leads to transformation and enhances tumorigenesis, and is a therapeutic target (85). HuR was recently found to interact with the 3′UTR of the eIF4E mRNA, stabilizing the eIF4E mRNA and increasing eIF4E expression (86). As postulated by Topisirovic and co-workers, eIF4E and HuR could jointly upregulate the expression of proliferative proteins encoded by shared target mRNAs (86).

Enhancement of cell survival

Tumor cells develop under stress conditions, including reduced access to nutrients and growth factors and increased exposure to oxidative damage. In order to thrive, they must overcome death-causing signals, particularly those leading to apoptosis (programmed cell death). Apoptosis resistance is engendered by the expression of specific anti-apoptotic genes. As discussed below and elsewhere (39, 55, 87), HuR stabilizes and modulates the translation of numerous mRNAs and thereby promotes the expression of anti-apoptotic proteins and represses the expression of pro-apoptotic proteins. In addition, HuR was recently reported to regulate the splicing of the apoptosis-promoting receptor Fas; the resulting exclusion of an exon rendered a soluble Fas isoform that prevented cell death (19). It is important to note that under conditions of prolonged or severe stress, HuR can exert a pro-apoptotic influence instead (reviewed in 87).

Prothymosin α (ProTα)

ProTα protects against apoptosis by inhibiting the formation of the apoptosome, a cytosolic macromolecular complex that assembles in cells committed to apoptotic death. The apoptosome activates caspase-9, which in turn activates effector caspases culminating in apoptotic cell death (88). In this capacity, ProTα plays a key role in cell survival and is highly expressed in several cancers (88, 89). HuR was shown to promote ProTα expression by interacting with the 3′UTR of ProTα mRNA and enhancing its translation (55). In turn, the HuR-mediated increase in ProTα expression protected cervical carcinoma cells against irradiation with short-wavelength ultraviolet light (UVC) (55).

B-cell lymphoma 2 (Bcl-2)

The proto-oncogene Bcl-2 is expressed in many cancers and is a major anti-apoptotic protein, inhibiting the release of cytochrome c from the mitochondria (90). In leukemia and carcinoma lines, HuR was shown to bind the Bcl-2 mRNA, stabilized it, and promoted Bcl-2 translation, leading to a significant increase in Bcl-2 abundance (39, 91).

Myeloid cell leukemia-1 (Mcl-1)

Mcl-1 is highly similar to Bcl-2, both structurally and functionally, and is also highly expressed in several cancer cells (92). Although molecular details of the influence of HuR on Mcl-1 expression are unknown, in cervical carcinoma cells HuR interacts with the Mcl-1 mRNA and HuR downregulation lowers Mcl-1 mRNA and protein abundance (39).

Sirtuin 1 (SIRT1)

SIRT1 promotes cell survival by deacetylating and thereby suppressing the activities of pro-apoptotic proteins such as p53, Foxo-3a and Ku70 (reviewed in 93). SIRT1 is highly expressed in different cancers including colon, prostate, and cervical cancers and has been ascribed a number of pro-tumor functions (94). HuR was found to interact with the SIRT1 3′UTR, enhanced SIRT1 mRNA stability, and thereby increased SIRT1 levels (14). In turn, cells expressing high SIRT1 showed heightened resistance to apoptosis triggered by various damaging agents (14, 95).

p21WAF1

A universal inhibitor of cdks, p21 reduces cell cycle progression and proliferation (96). p21 has been proposed to be pro-oncogenic by its ability to inhibit apoptosis in response to stress signals, through both p53-dependent and p53-independent mechanisms (97, 98). Accordingly, decreased p21 expression following preoperative chemoradiotherapy for rectal cancer was associated with improved disease-free survival (99). HuR interacts with the 3′UTR of the p21 mRNA (32); after exposure to UVC or ionizing irradiation, HuR increased the stability of p21 mRNA and elevated p21 expression (17, 32). These findings suggest that HuR could enhance p21 expression during the course of radiotherapy, conferring increased resistance to the cancer cell.

Mdm2

The Mdm2 p53-binding protein promotes cell survival. It inactivates p53 function by binding to its transactivation domain and targeting it for proteasome-mediated proteolysis. Recently, HuR was reported to interact with the Mdm-2 mRNA, although the specific region of binding was not determined (100). In inducible HuR-null mice, Mdm-2 mRNA stability was markedly reduced, leading to the reduced expression of Mdm-2, which the authors linked to widespread apoptosis in the intestinal epithelium, loss of intestinal villi, and lethality by 10 days after induction of the HuR-null phenotype (100).

c-Myc

Among many complex cellular functions, the c-Myc transcription factor is potently pro-apoptotic (101, 102). Aknown HuR target (103), the c-Myc mRNA was recently shown to be subject to translational repression by HuR; this process required the recruitment of the microRNA let-7 to a site adjacent to the HuR-binding region on the c-Myc 3′UTR (53). Together with the enhanced expression of the anti-apoptotic proteins listed above, the repression of the pro-apoptotic c-Myc solidifies a role of HuR as a strong anti-apoptotic factor.

Elevation of local angiogenesis

In order to expand, tumor cells need to develop local vasculature. This process, termed angiogenesis, permits the delivery of nutrients and oxygen and allows tumors to thrive (104). Numerous signals can promote or inhibit angiogenesis. Below we include examples of HuR promoting the expression of pro-angiogenic factors and halting the production of anti-angiogenic factors.

Hypoxia-inducible factor alpha (HIF-1α)

The transcription factor HIF-1α is robustly expressed in cells growing in the presence of low oxygen concentrations (hypoxia). Under these conditions, HIF-1α plays a key role in the transcriptional activation of several genes that are essential for the cell’s adaptation to hypoxia (105, 106). Several studies have reported increased HIF-1α abundance in tumors, a correlation between HIF-1α levels and aggressive cancer phenotypes, and a role for HIF-1α as a potential target for cancer therapy (107, 108). Among the many factors that associate with HIF-1α mRNA and modulate its expression, HuR interacts with the HIF-1α 5′UTR and promotes HIF-1α translation, and with the HIF-1α 3′UTR and stabilizes it (83,109111). Among other cancer-related functions, HIF-1α is potently cytoprotective (112).

Vascular endothelial growth factor (VEGF)

VEGF enhances cancer development by promoting cell growth, angiogenesis, proliferation, and migration (113116). VEGF is highly expressed in tumor cells growing in hypoxic conditions. The VEGF mRNA was one of the first HuR targets identified (30); binding of HuR to the VEGF 3′UTR increased VEGF mRNA stability and VEGF production in cultured cells and in tumors (30, 117). As the VEGF gene is also a transcriptional target of HIF-1α, increases in HuR both enhance the transcription of VEGF mRNA (via the heightened abundance of HIF-1α) and stabilize the VEGF mRNA, potently elevating VEGF production.

Cyclooxygenase-2 (COX-2)

COX-2 is highly expressed in many cancers, associated with increased VEGF levels and angiogenesis (118, 119). Although COX-2 plays a major role in mediating inflammation, its inhibitors are also effective anticancer drugs which function by blocking angiogenesis and tumor proliferation (119, 120). HuR binds to an AU-rich sequence in the COX-2 3′UTR, stabilizes the COX-2 mRNA, and increases COX-2 biosynthesis (121). Colon, ovarian, and prostate cancer cells with elevated cytoplasmic HuR levels also expressed higher levels of COX-2 and in one recent report, HuR was shown to regulate COX-2 levels during colon carcinogenesis (122125). The same correlation between HuR and COX-2 levels was seen in human mesothelioma, where it was further established that overall survival was strongly influenced by the subcellular localization of HuR (126).

Thrombospondin

Expression of the oncogene and inhibitor of angiogenesis thrombospondin (TSP1) is often lost in tumors (127, 128). HuR associates with the TSP1 3′UTR and enhances its translation (62). Interestingly, in a model of breast cancer progression, the association of HuR with TSP1 mRNA was progressively reduced as the cells displayed increasingly tumorigenic phenotypes. In highly malignant cells, this reduced interaction resulted in the diminished expression of TSP1; in turn, reduced TSP1 levels enhanced the pro-angiogenic phenotype of the cancer cells (62). Considering that HuR increases pro-angiogenic factors (VEGF, HIF-1α, and COX-2), the inhibition of TSP1 synthesis by HuR’s dissociation from TSP1 mRNA underscores HuR’s efficient coordination of an angiogenic program in cancer cells.

Reduced immune recognition

Surveillance by the immune system leads to the elimination of tumor cells. Therefore, cancer cells have adopted mechanisms to avoid recognition by immune cells. Despite the scarcity of examples to-date of HuR regulating proteins to escape immune surveillance, it is worth mentioning that HuR-mediated increase in MKP-1 levels in immune cells could help to accomplish the same phenotype, as MKP-1 potently suppresses immune function (129, 130). As reported in cervical carcinoma cells, HuR was interacted with the MKP-1 mRNA, stabilized it, and enhanced its translation following exposure to oxidative stress (58).

Transforming growth factor β (TGF-β)

The cytokine TGF-β is involved in cell proliferation, differentiation, and apoptosis (131). Although TGF-β can suppress the development of early-stage tumors, it promotes late-stage tumor growth by stimulating proliferation, invasiveness, and metastasis (132, 133). Some studies have linked the tumorigenic influence of TGF-β to the fact that it enables tumor cells to escape immune recognition (134, 135). HuR binds with high affinity the 3′UTR of TGF-β mRNA and was shown to regulate its expression post-transcriptionally in malignant brain tumors (136); this function could be important to evade immune recognition and allow cancer development.

Invasion and metastasis

Eventually, tumor cells will invade adjacent tissues and colonize distant tissues. These complex processes implicate changes in the interaction of the cancer cell with its local environment and the increased function of extracellular proteases.

Snail

Overexpression of the transcription factor snail is responsible for the induction of the epithelial-to-mesenchymal transition (EMT), through the repression of epithelial factors such as E-cadherin and cytokeratins, and the induction of mesenchymal proteins like fibronectin and metalloproteases (137). HuR was found to interact with the 3′UTR of snail mRNA and stabilized it following oxidative stress (138).

Matrix metalloproteinase 9 (MMP-9)

MMPs are metallopeptidases capable of cleaving most extracellular matrix (ECM) substrates, including collagens, laminin, fibronectin, vitronectin and proteoglycans in both physiological and pathological conditions (139, 140). MMP-9 is highly expressed in different cancers and is normally associated with high levels of invasion and/or metastasis of cervical, colorectal, gastric, pancreatic, breast, and oral cancers, as well as in glioma and skin tumors (141144). In MMP-9-null mice, tumors that mimic human recurrent primary tumors did not grow; likewise, inhibition of MMP-9 limited or inhibited local tumor invasion and metastasis (144, 145). HuR was reported to bind to the 3′UTR of MMP-9 mRNA and stabilized it, an effect that was enhanced by extracellular ATP and was suppressed by nitric oxide-triggered reduction in HuR levels (146, 147).

Urokinase A (uPA) and uPA-receptor (uPAR)

The urokinase-type plasminogen activator (uPA) and uPA receptor (uPAR) are members of a serine proteinase system that participates in ECM degradation, thereby affecting cell adhesion, invasion, and metastasis (148). uPA and uPAR are overexpressed in several malignant tumors, associated with poor disease outcome (148, 149). In an interesting example of coordinate regulation of functionally related proteins, HuR interacted with the 3′UTRs of both uPA and uPAR mRNAs, increased their stabilities, and enhanced expression of the encoded proteins (36). In this manner, HuR stimulates the uPA/uPAR signaling pathway leading to ECM degradation and facilitating tumor invasion and metastasis (150).

HuR EXPRESSION IN CANCER

The discovery that HuR was broadly elevated in cancer tissues compared with the corresponding non-cancer tissues (60) has been complemented by the analysis of HuR levels in specific cancers. To-date, the bulk of the studies on HuR in cancer have examined correlations between HuR abundance in cancer tissues and the stage and grade of the cancer. Recent studies have begun to assess the possible diagnostic, prognostic, and therapeutic value of HuR in cancer.

Breast cancer

Increased cytoplasmic HuR levels were found to be associated with high-grade, invasive ductal breast carcinoma, as well as with poor outcome, large tumor size, and poor survival rates (151). In breast cancer cells, HuR increased expression of Cyclin E1, IL-8, estrogen receptor, TSP1, and c-fms (62, 74, 152154). HuR was found to be more abundant in the cytoplasm after treatment with tamoxifen, suggesting that HuR could modulate the sensitivity to tamoxifen (155). Interestingly, in human breast epithelial cells, HuR was found to interact with the Wnt5a 3′UTR and repressed the translation of Wnt5a, a protein that inhibits tumor growth (52). HuR also associated with the BRCA1 3′UTR and reduced BRCA1 expression, although the molecular details of this influence were not studied (156). HuR was identified as an important prognostic factor in a subset of breast cancers, where it constituted an independent marker of reduced patient survival (157).

Colon cancer

Increased expression of HuR in colon cancer tissues promoted the expression of COX-2 and VEGF (121, 123), while the cytoplasmic abundance of HuR was associated with COX-2 expression levels and with high tumor stage (158). Importantly, overexpression of HuR increased the growth of colon cancer cells in a nude mouse xenograft model (60).

Ovarian cancer

HuR was found in the cytoplasm of ~50% of serous-type ovarian carcinoma, associated with elevated COX-2 expression, high grade, and poor patient prognosis (125). HuR was also found to interact with the 3′UTR of the ARHI/DRAS3 mRNA, which encodes a tumor suppressor protein, and likely increases ARHI expression (159). However, in ovarian cancer cells, HuR showed diminished interaction with ARHI mRNA, associated with the reduced expression of AHRI (159). One study examining the distribution of HuR in ~100 ovarian carcinomas revealed that HuR was primarily nuclear, but was found in the cytoplasm of 45% of ovarian carcinomas; this localization was associated with increased COX-2 expression, tumor grade, and mitotic activity, and with reduced overall patient survival (160). On the other hand, a large screen of ovarian carcinomas revealed an association between nuclear HuR and invasive cancer, high tumor grade, and decreased disease-free survival, indicating that nuclear HuR may also play a key role in ovarian tumorigenesis (161).

Prostate cancer

The analysis of HuR in over 100 primary prostate carcinoma samples revealed that HuR was predominantly nuclear in normal prostate glands but showed an elevated cytoplasmic presence in prostate carcinoma; the shift in localization that was linked to the levels of prostate-specific antigen (PSA) (162). Patients whose tumors showed elevated cytoplasmic HuR expressed higher COX-2, had shorter disease-free survival times, and had adverse prognosis, supporting the view that cytoplasmic HuR promotes prostate tumor development and relapse (124, 162).

Pancreatic cancer

In a recent study, elevated abundance of cytoplasmic HuR in pancreatic ductal adenocarcinoma (PDA) was associated with a 7-fold lower risk of patient mortality (163). The authors linked the cytoplasmic levels of HuR with binding of HuR to the deoxycytidine kinase (dCK) mRNA leading to its stabilization and the enhanced expression of dCK. This observation was significant because dCK metabolizes, and thereby activates, gemcitabine, a major component of a common chemotherapeutic regimen for PDA. Accordingly, Costantino and colleagues proposed that the presence of elevated HuR enhances the efficacy of gemcitabine in PDA and improves patient outcome (163).

Oral cancer

HuR expression levels were determined in salivary pleomorphic adenoma and salivary mucoepidermoid carcinoma. One third of pleomorphic adenomas and most mucoepidermoid carcinomas exhibited high cytoplasmic HuR levels; in the latter group, COX-2 expression was also elevated. This study suggests that in salivary carcinoma, cytoplasmic HuR levels correlate with COX-2 expression and that COX-2 and cytoplasmic HuR immunoreactivity could be used to evaluate malignancy in the salivary glands (164). A comparison between oral cancer and normal cells in culture revealed that HuR was nuclear in normal cells, but it was abundantly cytoplasmic in the oral cancer lines (165). Moreover, while inhibition of CRM1 reportedly blocked HuR export to the cytoplasm, in oral cancer cells HuR was readily exported to the cytoplasm despite CRM1 inhibition. These findings suggest that the cytoplasmic export of HuR is different in oral carcinoma cells compared with normal cells (165). Whether HuR nuclear export is also aberrant in other tumor types awaits further study.

Other cancers

In gastric cancer, HuR concentration also correlated with COX-2 expression and with poor survival rates, suggesting that HuR may also enhance gastric carcinogenesis (166, 167). Similarly, a comparison of primary Merkel cell carcinoma (MCC) and non-neoplastic skin showed that cytoplasmic HuR was higher in a subset of MMC tumors than in non-neoplastic skin, supporting a role for HuR in MMC carcinogenesis (168). In tumors of the central nervous system (CNS), HuR overexpression was linked to the enhanced expression of COX-2, VEGF, TGF-β and other factors involved in CNS tumor proliferation and angiogenesis (136).

CONCLUDING REMARKS

HuR interacts with and regulates many mRNAs encoding cancer-related proteins (11, 12, 41, 60). By modulating their expression in a coordinated manner, HuR can have a profound impact on multiple phenotypic traits central to tumorigenesis. The heightened expression of HuR, particularly cytoplasmic HuR, in virtually all cancers examined suggests that HuR could be a useful diagnostic marker. However, it is important to measure HuR protein levels instead of HuR mRNA levels, as the latter do not change markedly between normal and cancer tissues (9, 169). Why HuR protein levels change with cancer without changes in HuR mRNA levels is not fully understood, but may be linked to the stability of the protein itself (18) or to the translational repression of HuR by microRNAs such as miR-519 and miR-125a, which inhibit HuR translation without affecting HuR mRNA levels (169, 170). Similarly, the subcellular localization of HuR and its ability to bind mRNAs strongly influences the expression of target mRNAs. These facets of HuR are modulated by several post-translational modifications and transport machineries; virtually all of the HuR-modifying enzymes (Chk2, PKC, CARM1, Cdk1, p38, caspases) have been implicated in cancer-related processes (Fig. 1). Further studies to understand the complex mechanisms that regulate HuR abundance, localization, and binding to target transcripts in cancer cells are warranted.

Given the extensive catalog of genes that HuR controls, and HuR’s positive influence on angiogenesis, proliferation, survival, invasion, etc, strategies to reduce HuR expression or inhibit its function could be successful in reducing tumor progression. Silencing approaches using small interfering (si)iRNA or microRNAs appear to be effective in cultured cells and might be attempted in tumors. In addition, small chemical inhibitors of HuR have been reported, but their usefulness in organisms also remain untested (171). However, it is important to note that most studies on HuR and cancer have not examined how HuR might affect anti-cancer therapy. In the Costantino report mentioned above, the elevated presence of cytoplasmic HuR in pancreatic cancer cells paradoxically correlated with better prognosis in patients treated with the standard drug of choice, gemcitabine; that HuR increased the expression of deoxycytidine kinase, which metabolizes and thus activates gemcitabine, helped to explain why elevated HuR was associated with positive response to therapy. Similarly, low HuR levels were associated with high risk of breast cancer recurrence, although the specific mediators of this effect were not identified (172). Therefore, interventions to reduce HuR function should be devised carefully. In some cases, the elevated presence of HuR may be advantageous for therapy and could be exploited to that end.

While the use of cultured cells has advanced greatly our understanding of HuR function and influence on cancer-associated proteins, major efforts must now focus on mammalian models of carcinogenesis (173). Mouse HuR-null thymocytes showed aberrant cell division cycle, activation, selection, and survival (174). A role for HuR in the establishment of a physiologic thymocyte pool was confirmed in another mouse model with inducible global HuR-null phenotype, which showed widespread death of progenitor cells in hematopoietic organs and in the intestinal epithelium (100). While the mouse phenotypes agree with HuR’s roles in proliferation and survival, more studies are needed to elucidate if an HuR-null status affects tumorigenesis. Transgenic mice overexpressing HuR will also be highly informative in providing insight into the role of HuR in cancer. With the knowledge gained thus far of HuR-regulated gene expression programs, the stage is ready for direct assessment of HuR’s diagnostic, prognostic, and therapeutic potential in human cancer.

Acknowledgments

K.A. and M.G. are supported by the National Institute on Aging-Intramural Research Program, National Institutes of Health.

Abbreviations

AMPK

AMP-activated protein kinase

Bcl-2

B-cell lymphoma 2

Cdk1

cyclin-dependent kinase 1

Chk2

checkpoint kinase 2

CNS

central nervous system

COX-2

cyclooxygenase-2

CR

coding region

dCK

deoxycytidine kinase

eIF4E

eukaryotic translation initiation factor 4E

Elav

embryonic lethal abnormal vision

GM-CSF

granulocyte/macrophage-colony-stimulating factor

HIF

hypoxia-inducible factor

HNS

HuR nucleocytoplasmic shuttling sequence

IGF-IR

type-I insulin-like growth factor receptor

IL

interleukin

iNOS

inducible nitric oxide synthase

IRES

internal ribosome entry site

MAPK

mitogen-activated protein kinase

Mcl-1

myeloid cell leukemia-1

MKP-1

MAPK phosphatase-1

MMP-9

matrix metalloproteinase 9

PKC

protein kinase C

PSA

prostate-specific antigen

RBP

RNA-binding protein

RRM

RNA recognition motif

SIRT1

sirtuin 1

TGF-β

transforming growth factor β

TNF

tumor necrosis factor

TSP1

thrombospondin

uPA

urokinase-type plasminogen activator

uPAR

uPA receptor

UTR

untranslated region

VEGF

vascular endothelial growth factor

BIBLIOGRAPHY

  • 1.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 2.Sager R. Expression genetics in cancer: shifting the focus from DNA to RNA. Proc Natl Acad Sci USA. 1997;94:952–955. doi: 10.1073/pnas.94.3.952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.López de Silanes I, Quesada MP, Esteller M. Aberrant regulation of messenger RNA 3′-untranslated region in human cancer. Cell Oncol. 2007;29:1–17. doi: 10.1155/2007/586139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lukong KE, Chang KW, Khandjian EW, Richard S. RNA-binding proteins in human genetic disease. Trends Genet. 2008;24:416–425. doi: 10.1016/j.tig.2008.05.004. [DOI] [PubMed] [Google Scholar]
  • 5.Kim MY, Hur J, Jeong S. Emerging roles of RNA and RNA-binding protein network in cancer cells. BMB Rep. 2009;42:125–130. doi: 10.5483/bmbrep.2009.42.3.125. [DOI] [PubMed] [Google Scholar]
  • 6.Moore MJ. From birth to death: the complex lives of eukaryotic mRNAs. Science. 2005;309:1514–1518. doi: 10.1126/science.1111443. [DOI] [PubMed] [Google Scholar]
  • 7.Keene JD. RNA regulons: coordination of post-transcriptional events. Nat Rev Genet. 2007;8:533–543. doi: 10.1038/nrg2111. [DOI] [PubMed] [Google Scholar]
  • 8.Gouble A, Grazide S, Meggetto F, Mercier P, Delsol G, Morello D. A new player in oncogenesis: AUF1/hnRNPD overexpression leads to tumorigenesis in transgenic mice. Cancer Res. 2002;62:1489–1495. [PubMed] [Google Scholar]
  • 9.Brennan SE, Kuwano Y, Alkharouf N, Blackshear PJ, Gorospe M, Wilson GM. The mRNA-destabilizing protein tristetraprolin is suppressed in many cancers, altering tumorigenic phenotypes and patient prognosis. Cancer Res. 2009;69:5168–5176. doi: 10.1158/0008-5472.CAN-08-4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Szabo A, Dalmau J, Manley G, Rosenfeld M, Wong E, Henson J, Posner JB, Furneaux HM. HuD, a paraneoplastic encephalomyelitis antigen, contains RNA-binding domains and is homologous to Elav and Sex-lethal. Cell. 1991;67:325–333. doi: 10.1016/0092-8674(91)90184-z. [DOI] [PubMed] [Google Scholar]
  • 11.Hinman MN, Lou H. Diverse molecular functions of Hu proteins. Cell Mol Life Sci. 2008;65:3168–3181. doi: 10.1007/s00018-008-8252-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Masuda K, Marasa B, Martindale JL, Halushka MK, Gorospe M. Tissue- and age-dependent expression of RNA-binding proteins that influence mRNA turnover and translation. Aging. 2009 doi: 10.18632/aging.100073. in Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.López de Silanes I, Zhan M, Lal A, Yang X, Gorospe M. Identification of a target RNA motif for RNA-binding protein HuR. Proc Natl Acad Sci USA. 2004;101:2987–2992. doi: 10.1073/pnas.0306453101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Abdelmohsen K, Pullmann P, Jr, Lal A, Kim HH, Galban S, Yang X, Blethrow JD, Walker M, Shubert J, Gillespie DA, Furneaux H, Gorospe M. Phosphorylation of HuR by Chk2 regulates SIRT1 expression. Mol Cell. 2007;25:543–557. doi: 10.1016/j.molcel.2007.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Doller A, Akool el-S, Huwiler A, Müller R, Radeke HH, Pfeilschifter J, Eberhardt W. Posttranslational modification of the AU-rich element binding protein HuR by protein kinase Cδ elicits angiotensin II-induced stabilization and nuclear export of cyclooxygenase 2 mRNA. Mol Cell Biol. 2008;28:2608–2625. doi: 10.1128/MCB.01530-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Doller A, Huwiler A, Müller R, Radeke HH, Pfeilschifter J, Eberhardt W. Protein kinase C alpha-dependent phosphorylation of the mRNA-stabilizing factor HuR: implications for posttranscriptional regulation of cyclooxygenase-2. Mol Biol Cell. 2007;18:2137–2148. doi: 10.1091/mbc.E06-09-0850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lafarga V, Cuadrado A, López de Silanes I, Bengoechea R, Fernandez-Capetillo O, Nebreda AR. p38 Mitogen-activated protein kinase- and HuR-dependent stabilization of p21(Cip1) mRNA mediates the G(1)/S checkpoint. Mol Cell Biol. 2009;29:4341–4351. doi: 10.1128/MCB.00210-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Abdelmohsen K, Srikantan S, Yang X, Lal A, Kim HH, Kuwano Y, Galban S, Becker KG, Kamara D, de Cabo R, Gorospe M. Ubiquitin-mediated proteolysis of HuR by heat shock. EMBO J. 2009;28:1271–1282. doi: 10.1038/emboj.2009.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Izquierdo JM. Hu antigen R (HuR) functions as an alternative pre-mRNA splicing regulator of Fas apoptosis-promoting receptor on exon definition. J Biol Chem. 2008;283:19077–19084. doi: 10.1074/jbc.M800017200. [DOI] [PubMed] [Google Scholar]
  • 20.Gallouzi IE, Steitz JA. Delineation of mRNA export pathways by the use of cell-permeable peptides. Science. 2001;294:1895–1901. doi: 10.1126/science.1064693. [DOI] [PubMed] [Google Scholar]
  • 21.Fan XC, Steitz JA. HNS, a nuclear-cytoplasmic shuttling sequence in HuR. Proc Natl Acad Sci USA. 1998;95:15293–15298. doi: 10.1073/pnas.95.26.15293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Güttinger S, Mühlhäusser P, Koller-Eichhorn R, Brennecke J, Kutay U. Transportin2 functions as importin and mediates nuclear import of HuR. Proc Natl Acad Sci USA. 2004;101:2918–23. doi: 10.1073/pnas.0400342101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rebane A, Aab A, Steitz JA. Transportins 1 and 2 are redundant nuclear import factors for hnRNP A1 and HuR. RNA. 2004;10:590–599. doi: 10.1261/rna.5224304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang W, Fan J, Yang X, Fürer-Galban S, López de Silanes I, von Kobbe C, Guo J, Georas SN, Foufelle F, Hardie DG, Carling D, Gorospe M. AMP-activated kinase regulates cytoplasmic HuR. Mol Cell Biol. 2002;22:3425–3436. doi: 10.1128/MCB.22.10.3425-3436.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kim HH, Abdelmohsen K, Lal A, Pullmann R, Jr, Yang X, Galban S, Srikantan S, Martindale JL, Blethrow J, Shokat KM, Gorospe M. Nuclear HuR accumulation through phosphorylation by Cdk1. Genes Dev. 2008;22:1804–1815. doi: 10.1101/gad.1645808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kim HH, Yang X, Kuwano Y, Gorospe M. Modification at HuR(S242) alters HuR localization and proliferative influence. Cell Cycle. 2008;7:3371–3377. doi: 10.4161/cc.7.21.6895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kim HH, Gorospe M. Phosphorylated HuR shuttles in cycles. Cell Cycle. 2008;7:3124–3126. doi: 10.4161/cc.7.20.6884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Li H, Park S, Kilburn B, Jelinek MA, Henschen-Edman A, Aswad DW, Stallcup MR, Laird-Offringa IA. Lipopolysaccharide-induced methylation of HuR, an mRNA-stabilizing protein, by CARM1. Coactivator-associated arginine methyltransferase. J Biol Chem. 2002;277:44623–44630. doi: 10.1074/jbc.M206187200. [DOI] [PubMed] [Google Scholar]
  • 29.Mazroui R, Di Marco S, Clair E, von Roretz C, Tenenbaum SA, Keene JD, Saleh M, Gallouzi IE. Caspase-mediated cleavage of HuR in the cytoplasm contributes to pp32/PHAP-I regulation of apoptosis. J Cell Biol. 2008;180:113–127. doi: 10.1083/jcb.200709030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem. 1998;273:6417–6423. doi: 10.1074/jbc.273.11.6417. [DOI] [PubMed] [Google Scholar]
  • 31.Wang W, Caldwell MC, Lin S, Furneaux H, Gorospe M. HuR regulates cyclin A and cyclin B1 mRNA stability during cell proliferation. EMBO J. 2000;19:2340–2350. doi: 10.1093/emboj/19.10.2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang W, Furneaux H, Cheng H, Caldwell MC, Hutter D, Liu Y, Holbrook N, Gorospe M. HuR regulates p21 mRNA stabilization by UV light. Mol Cell Biol. 2000;20:760–769. doi: 10.1128/mcb.20.3.760-769.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ming XF, Stoecklin G, Lu M, Looser R, Moroni C. Parallel and independent regulation of interleukin-3 mRNA turnover by phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase. Mol Cell Biol. 2001;21:5778–5789. doi: 10.1128/MCB.21.17.5778-5789.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen CY, Xu N, Shyu AB. Highly selective actions of HuR in antagonizing AU-rich element-mediated mRNA destabilization. Mol Cell Biol. 2002;22:7268–7278. doi: 10.1128/MCB.22.20.7268-7278.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sengupta S, Jang BC, Wu MT, Paik JH, Furneaux H, Hla T. The RNA-binding protein HuR regulates the expression of cyclooxygenase-2. J Biol Chem. 2003;278:25227–2533. doi: 10.1074/jbc.M301813200. [DOI] [PubMed] [Google Scholar]
  • 36.Tran H, Maurer F, Nagamine Y. Stabilization of urokinase and urokinase receptor mRNAs by HuR is linked to its cytoplasmic accumulation induced by activated mitogen-activated protein kinase-activated protein kinase 2. Mol Cell Biol. 2003;23:7177–7188. doi: 10.1128/MCB.23.20.7177-7188.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lal A, Mazan-Mamczarz K, Kawai T, Yang X, Martindale JL, Gorospe M. Concurrent versus individual binding of HuR and AUF1 to common labile target mRNAs. EMBO J. 2004;23:3092–3102. doi: 10.1038/sj.emboj.7600305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Song IS, Tatebe S, Dai W, Kuo MT. Delayed mechanism for induction of gamma-glutamylcysteine synthetase heavy subunit mRNA stability by oxidative stress involving p38 mitogen-activated protein kinase signaling. J Biol Chem. 2005;280:28230–28240. doi: 10.1074/jbc.M413103200. [DOI] [PubMed] [Google Scholar]
  • 39.Abdelmohsen K, Lal A, Kim HH, Gorospe M. Posttranscriptional orchestration of an anti-apoptotic program by HuR. Cell Cycle. 2007;6:1288–1292. doi: 10.4161/cc.6.11.4299. [DOI] [PubMed] [Google Scholar]
  • 40.Brennan CM, Steitz JA. HuR and mRNA stability. Cell Mol Life Sci. 2001;58:266–277. doi: 10.1007/PL00000854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Abdelmohsen K, Kuwano Y, Kim HH, Gorospe M. Posttranscriptional gene regulation by RNA-binding proteins during oxidative stress: implications for cellular senescence. Biol Chem. 2008;389:243–255. doi: 10.1515/BC.2008.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chen CY, Gherzi R, Ong SE, Chan EL, Raijmakers R, Pruijn GJ, Stoecklin G, Moroni C, Mann M, Karin M. AU binding proteins recruit the exosome to degrade ARE-containing mRNAs. Cell. 2001;107:451–464. doi: 10.1016/s0092-8674(01)00578-5. [DOI] [PubMed] [Google Scholar]
  • 43.Butler JS. The yin and yang of the exosome. Trends Cell Biol. 2002;12:90–96. doi: 10.1016/s0962-8924(01)02225-5. [DOI] [PubMed] [Google Scholar]
  • 44.Mukherjee D, Gao M, O’Connor JP, Raijmakers R, Pruijn G, Lutz CS, Wilusz J. The mammalian exosome mediates the efficient degradation of mRNAs that contain AU-rich elements. EMBO J. 2002;21:165–174. doi: 10.1093/emboj/21.1.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gherzi R, Lee KY, Briata P, Wegmuller D, Moroni C, Karin M, Chen CY. A KH-domain RNA-binding protein, KSRP, promotes ARE-directed mRNA turnover by recruiting the degradation machinery. Mol Cell. 2004;14:571–583. doi: 10.1016/j.molcel.2004.05.002. [DOI] [PubMed] [Google Scholar]
  • 46.Ingelfinger D, Arndt-Jovin DJ, Luhrmann R, Achsel T. The human LSm1-7 proteins colocalize with the mRNA-degrading enzymes Dcp1/2 and Xrnl in distinct cytoplasmic foci. RNA. 2002;8:1489–1501. [PMC free article] [PubMed] [Google Scholar]
  • 47.Eystathioy T, Jakymiw A, Chan EK, Seraphin B, Cougot N, Fritzler MJ. The GW182 protein colocalizes with mRNA degradation associated proteins hDcp1 and hLSm4 in cytoplasmic GW bodies. RNA. 2003;9:1171–1173. doi: 10.1261/rna.5810203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cougot N, Babajko S, Seraphin B. Cytoplasmic foci are sites of mRNA decay in human cells. J Cell Biol. 2004;165:31–40. doi: 10.1083/jcb.200309008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kedersha N, Stoecklin G, Ayodele M, Yacono P, Lykke- Andersen J, Fitzler MJ, Scheuner D, Kaufman RJ, Golan DE, Anderson P. Stress granules and processing bodies are dynamically linked sites of mRNP remodeling. J Cell Biol. 2005;169:871–884. doi: 10.1083/jcb.200502088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kullmann M, Gopfert U, Siewe B, Hengst L. ELAV/Hu proteins inhibit p27 translation via an IRES element in the p27 5′UTR. Genes Dev. 2002;16:3087–3099. doi: 10.1101/gad.248902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Meng Z, King PH, Nabors LB, Jackson NL, Chen CY, Emanuel PD, Blume SW. The ELAV RNA-stability factor HuR binds the 59-untranslated region of the human IGF-IR transcript and differentially represses capdependent and IRES-mediated translation. Nucleic Acids Res. 2005;33:2962–2979. doi: 10.1093/nar/gki603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Leandersson K, Riesbeck K, Andersson T. Wnt-5a mRNA translation is suppressed by the Elav-like protein HuR in human breast epithelial cells. Nucleic Acids Res. 2006;34:3988–3999. doi: 10.1093/nar/gkl571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kim HH, Kuwano Y, Srikantan S, Lee EK, Martindale JL, Gorospe M. HuR recruits let-7/RISC to repress c-Myc expression. Genes Dev. 2009;23:1743–1748. doi: 10.1101/gad.1812509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mazan-Mamczarz K, Galban S, López de Silanes I, Martindale JL, Atasoy U, Keene JD, Gorospe M. RNA-binding protein HuR enhances p53 translation in response to ultraviolet light irradiation. Proc Natl Acad Sci USA. 2003;100:8354–8359. doi: 10.1073/pnas.1432104100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lal A, Kawai T, Yang X, Mazan-Mamczarz K, Gorospe M. Antiapoptotic function of RNA-binding protein HuR effected through prothymosin alpha. EMBO J. 2005;24:1852–1862. doi: 10.1038/sj.emboj.7600661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Bhattacharyya SN, Habermacher R, Martine U, Closs EI, Filipowicz W. Relief of microRNA-mediated translational repression in human cells subjected to stress. Cell. 2006;125:1111–1124. doi: 10.1016/j.cell.2006.04.031. [DOI] [PubMed] [Google Scholar]
  • 57.Kawai T, Lal A, Yang X, Galban S, Mazan-Mamczarz K, Gorospe M. Translational control of cytochrome c by RNA-binding proteins TIA-1 and HuR. Mol Cell Biol. 2006;26:3295–3307. doi: 10.1128/MCB.26.8.3295-3307.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kuwano Y, Kim HH, Abdelmohsen K, Pullmann R, Jr, Martindale JL, Yang X, Gorospe M. MKP-1 mRNA stabilization and translational control by RNA-binding proteins HuR and NF90. Mol Cell Biol. 2008;28:4562–4575. doi: 10.1128/MCB.00165-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kuwano Y, Rabinovic A, Srikantan S, Gorospe M, Demple B. Analysis of nitric oxide-stabilized mRNAs in human fibroblasts reveals HuR-dependent heme oxygenase 1 upregulation. Mol Cell Biol. 2009;29:2622–2635. doi: 10.1128/MCB.01495-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.López de Silanes I, Fan J, Yang X, Zonderman AB, Potapova O, Pizer ES, Gorospe M. Role of the RNA-binding protein HuR in colon carcinogenesis. Oncogene. 2003;22:7146–7154. doi: 10.1038/sj.onc.1206862. [DOI] [PubMed] [Google Scholar]
  • 61.López de Silanes I, Lal A, Gorospe M. HuR: post-transcriptional paths to malignancy. RNA Biol. 2005;2:11–13. doi: 10.4161/rna.2.1.1552. [DOI] [PubMed] [Google Scholar]
  • 62.Mazan-Mamczarz K, Hagner PR, Corl S, Srikantan S, Wood WH, Becker KG, Gorospe M, Keene JD, Levenson AS, Gartenhaus RB. Post-transcriptional gene regulation by HuR promotes a more tumorigenic phenotype. Oncogene. 2008;27:6151–6163. doi: 10.1038/onc.2008.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9:153–166. doi: 10.1038/nrc2602. [DOI] [PubMed] [Google Scholar]
  • 64.Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;9:729–740. doi: 10.1038/nrm2233. [DOI] [PubMed] [Google Scholar]
  • 65.Ohtani N, Mann DJ, Hara E. Cellular senescence: its role in tumor suppression and aging. Cancer Sci. 2009;100:792–797. doi: 10.1111/j.1349-7006.2009.01123.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang W, Yang X, Cristofalo VJ, Holbrook NJ, Gorospe M. Loss of HuR is linked to reduced expression of proliferative genes during replicative senescence. Mol Cell Biol. 2001;21:5889–5898. doi: 10.1128/MCB.21.17.5889-5898.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hosokawa Y, Arnold A. Mechanism of cyclin D1 (CCND1, PRAD1) overexpression in human cancer cells: analysis of allele-specific expression. Genes Chromosomes Cancer. 1998;22:66–71. doi: 10.1002/(sici)1098-2264(199805)22:1<66::aid-gcc9>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 68.Musgrove EA, Lee CS, Buckley MF, Sutherland RL. Cyclin D1 induction in breast cancer cells shortens G1 and is sufficient for cells arrested in G1 to complete the cell cycle. Proc Natl Acad Sci USA. 1994;91:8022–8026. doi: 10.1073/pnas.91.17.8022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Knudsen KE, Diehl JA, Haiman CA, Knudsen ES. Cyclin D1: polymorphism, aberrant splicing and cancer risk. Oncogene. 2006;25:1620–1628. doi: 10.1038/sj.onc.1209371. [DOI] [PubMed] [Google Scholar]
  • 70.Akama Y, Yasui W, Yokozaki H, Kuniyasu H, Kitahara K, Ishikawa T, Tahara E. Frequent amplification of the cyclin E gene in human gastric carcinomas. Jpn J Cancer Res. 1995;86:617–621. doi: 10.1111/j.1349-7006.1995.tb02442.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bortner DM, Rosenberg MP. Induction of mammary gland hyperplasia and carcinomas in transgenic mice expressing human cyclin E. Mol Cell Biol. 1997;17:453–9. doi: 10.1128/mcb.17.1.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Keyomarsi K, Herliczek TW. The role of cyclin E in cell proliferation, development and cancer. Prog Cell Cycle Res. 1997;3:171–191. doi: 10.1007/978-1-4615-5371-7_14. [DOI] [PubMed] [Google Scholar]
  • 73.Hwang HC, Clurman BE. Cyclin E in normal and neoplastic cell cycles. Oncogene. 2005;24:2776–2786. doi: 10.1038/sj.onc.1208613. [DOI] [PubMed] [Google Scholar]
  • 74.Guo X, Hartley RS. HuR contributes to cyclin E1 deregulation in MCF-7 breast cancer cells. Cancer Res. 2006;66:7948–7956. doi: 10.1158/0008-5472.CAN-05-4362. [DOI] [PubMed] [Google Scholar]
  • 75.Guo X, Wu Y, Hartley RS. Cold-inducible RNA-binding protein contributes to human antigen R and cyclin E1 deregulation in breast cancer. Mol Carcinog. 2009 doi: 10.1002/mc.20582. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Yam CH, Fung TK, Poon RY. Cyclin A in cell cycle control and cancer. Cell Mol Life Sci. 2002;59:1317–1326. doi: 10.1007/s00018-002-8510-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cho NH, Kang S, Hong S, An HJ, Choi YH, Jeong GB, Choi HK. Elevation of cyclin B1, active cdc2, and HuR in cervical neoplasia with human papillomavirus type 18 infection. Cancer Lett. 2006;232:170–178. doi: 10.1016/j.canlet.2005.02.026. [DOI] [PubMed] [Google Scholar]
  • 78.Singhal S, Vachani A, Antin-Ozerkis D, Kaiser LR, Albelda SM. Prognostic implications of cell cycle, apoptosis, and angiogenesis biomarkers in non-small cell lung cancer: a review. Clin Cancer Res. 2005;11:3974–86. doi: 10.1158/1078-0432.CCR-04-2661. [DOI] [PubMed] [Google Scholar]
  • 79.Egloff AM, Vella LA, Finn OJ. Cyclin B1 and other cyclins as tumor antigens in immunosurveillance and immunotherapy of cancer. Cancer Res. 2006;66:6–9. doi: 10.1158/0008-5472.CAN-05-3389. [DOI] [PubMed] [Google Scholar]
  • 80.Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, Tempst P, Massagué J. Cloning of p27Kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals. Cell. 1994;78:59–66. doi: 10.1016/0092-8674(94)90572-x. [DOI] [PubMed] [Google Scholar]
  • 81.Wang X, Gorospe M, Huang Y, Holbrook NJ. p27Kip1 overexpression causes apoptotic death of mammalian cells. Oncogene. 1997;15:2991–2997. doi: 10.1038/sj.onc.1201450. [DOI] [PubMed] [Google Scholar]
  • 82.Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5:341–354. doi: 10.1038/nrc1609. [DOI] [PubMed] [Google Scholar]
  • 83.Sheflin LG, Zou AP, Spaulding SW. Androgens regulate the binding of endogenous HuR to the AU-rich 3′UTRs of HIF-1α and EGF mRNA. Biochem Biophys Res Commun. 2004;322:644–651. doi: 10.1016/j.bbrc.2004.07.173. [DOI] [PubMed] [Google Scholar]
  • 84.Sonenberg N. eIF4E, the mRNA cap-binding protein: from basic discovery to translational research. Biochem Cell Biol. 2008;86:178–183. doi: 10.1139/O08-034. [DOI] [PubMed] [Google Scholar]
  • 85.Graff JR, Konicek BW, Carter JH, Marcusson EG. Targeting the eukaryotic translation initiation factor 4E for cancer therapy. Cancer Res. 2008;68:631–634. doi: 10.1158/0008-5472.CAN-07-5635. [DOI] [PubMed] [Google Scholar]
  • 86.Topisirovic I, Siddiqui N, Borden KL. The eukaryotic translation initiation factor 4E (eIF4E) and HuR RNA operons collaboratively regulate the expression of survival and proliferative genes. Cell Cycle. 2009;8:960–961. [PubMed] [Google Scholar]
  • 87.Kuwano Y, Gallouzi IE, Gorospe M. Role of the RNA-binding protein HuR in Apoptosis and Apoptosome Function. The Apoptosome as an Emerging Therapeutic Tool. 2009 [Google Scholar]
  • 88.Jiang X, Kim HE, Shu H, Zhao Y, Zhang H, Kofron J, Donnelly J, Burns D, Ng SC, Rosenberg S, Wang X. Distinctive roles of PHAP proteins and prothymosin-alpha in a death regulatory pathway. Science. 2003;299:223–226. doi: 10.1126/science.1076807. [DOI] [PubMed] [Google Scholar]
  • 89.Letsas KP, Frangou-Lazaridis M. Surfing on prothymosin alpha proliferation and anti-apoptotic properties. Neoplasma. 2006;53:92–96. [PubMed] [Google Scholar]
  • 90.Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP, Wang X. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science. 1997;275:1129–1132. doi: 10.1126/science.275.5303.1129. [DOI] [PubMed] [Google Scholar]
  • 91.Ishimaru D, Ramalingam S, Sengupta TK, Bandyopadhyay S, Dellis S, Tholanikunnel BG, Fernandes DJ, Spicer EK. Regulation of Bcl-2 expression by HuR in HL60 leukemia cells and A431 carcinoma cells. Mol Cancer Res. 2009;7:1354–1166. doi: 10.1158/1541-7786.MCR-08-0476. [DOI] [PubMed] [Google Scholar]
  • 92.Michels J, Johnson PW, Packham G. Mcl-1. Int J Biochem Cell Biol. 2005;37:267–271. doi: 10.1016/j.biocel.2004.04.007. [DOI] [PubMed] [Google Scholar]
  • 93.Wojcik M, Mac-Marcjanek K, Wozniak LA. Physiological and pathophysiological functions of SIRT1. Mini Rev Med Chem. 2009;9:386–94. doi: 10.2174/1389557510909030386. [DOI] [PubMed] [Google Scholar]
  • 94.Liu T, Liu PY, Marshall GM. The critical role of the class III histone deacetylase SIRT1 in cancer. Cancer Res. 2009;69:1702–1705. doi: 10.1158/0008-5472.CAN-08-3365. [DOI] [PubMed] [Google Scholar]
  • 95.Brooks CL, Gu W. How does SIRT1 affect metabolism, senescence and cancer? Nat Rev Cancer. 2009;9:123–128. doi: 10.1038/nrc2562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ball KL. p21: structure and functions associated with cyclin-CDK binding. Prog Cell Cycle Res. 1997;3:125–134. doi: 10.1007/978-1-4615-5371-7_10. [DOI] [PubMed] [Google Scholar]
  • 97.Gorospe M, Wang X, Holbrook NJ. Functional role of p21 during the cellular response to stress. Gene Expr. 1999;7:377–385. [PMC free article] [PubMed] [Google Scholar]
  • 98.Gartel AL, Tyner AL. The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther. 2002;1:639–649. [PubMed] [Google Scholar]
  • 99.Rau B, Sturm I, Lage H, Berger S, Schneider U, Hauptmann S, Wust P, Riess H, Schlag PM, Dorken B, Daniel PT. Dynamic expression profile of p21WAF1/CIP1 and Ki-67 predicts survival in rectal carcinoma treated with preoperative radiochemotherapy. J Clin Oncol. 2003;21:3391–3401. doi: 10.1200/JCO.2003.07.077. [DOI] [PubMed] [Google Scholar]
  • 100.Ghosh M, Aguila HL, Michaud J, Ai Y, Wu MT, Hemmes A, Ristimaki A, Guo C, Furneaux H, Hla T. Essential role of the RNA-binding protein HuR in progenitor cell survival in mice. J Clin Invest. 2009 doi: 10.1172/JCI38263. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pelengaris S, Khan M, Evan GI. Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell. 2002;109:321–334. doi: 10.1016/s0092-8674(02)00738-9. [DOI] [PubMed] [Google Scholar]
  • 102.Hoffman B, Liebermann DA. Apoptotic signaling by c-MYC. Oncogene. 2008;27:6462–6472. doi: 10.1038/onc.2008.312. [DOI] [PubMed] [Google Scholar]
  • 103.Lafon I, Carballes F, Brewer G, Poiret M, Morello D. Developmental expression of AUF1 and HuR, two c-myc mRNA binding proteins. Oncogene. 1998;16:3413–3421. doi: 10.1038/sj.onc.1201895. [DOI] [PubMed] [Google Scholar]
  • 104.Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353–364. doi: 10.1016/s0092-8674(00)80108-7. [DOI] [PubMed] [Google Scholar]
  • 105.Semenza GL. HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol. 2000;88:1474–1480. doi: 10.1152/jappl.2000.88.4.1474. [DOI] [PubMed] [Google Scholar]
  • 106.Wenger RH, Stiehl DP, Camenisch G. Integration of oxygen signaling at the consensus HRE. Sci STKE. 2005;306:re12. doi: 10.1126/stke.3062005re12. [DOI] [PubMed] [Google Scholar]
  • 107.Maynard MA, Ohh M. The role of hypoxia-inducible factors in cancer. Cell Mol Life Sci. 2007;64:2170–2180. doi: 10.1007/s00018-007-7082-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Poon E, Harris AL, Ashcroft M. Targeting the hypoxia-inducible factor (HIF) pathway in cancer. Expert Rev Mol Med. 2009;11:e26. doi: 10.1017/S1462399409001173. [DOI] [PubMed] [Google Scholar]
  • 109.Masuda K, Abdelmohsen K, Gorospe M. RNA-binding proteins implicated in the hypoxic response. J Cell Mol Med. 2009 doi: 10.1111/j.1582-4934.2009.00842.x. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Galbán S, Gorospe M. Factors interacting with HIF-1α mRNA: novel therapeutic targets. Curr Pharm Des. 2009 doi: 10.2174/138161209789649376. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Galbán S, Kuwano Y, Pullmann R, Jr, Martindale JL, Kim HH, Lal A, Abdelmohsen K, Yang X, Dang Y, Liu JO, Lewis SM, Holcik M, Gorospe M. RNA-binding proteins HuR and PTB promote the translation of hypoxia-inducible factor 1alpha. Mol Cell Biol. 2008;28:93–107. doi: 10.1128/MCB.00973-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Moeller BJ, Cao Y, Li CY, Dewhirst MW. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5:429–441. doi: 10.1016/s1535-6108(04)00115-1. [DOI] [PubMed] [Google Scholar]
  • 113.Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13:9–22. [PubMed] [Google Scholar]
  • 114.Carmeliet P. VEGF as a key mediator of angiogenesis in cancer. Oncology. 2005;69 (Suppl 3):4–10. doi: 10.1159/000088478. [DOI] [PubMed] [Google Scholar]
  • 115.Veikkola T, Alitalo K. VEGFs, receptors and angiogenesis. Semin Cancer Biol. 1999;9:211–220. doi: 10.1006/scbi.1998.0091. [DOI] [PubMed] [Google Scholar]
  • 116.Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407:242–248. doi: 10.1038/35025215. [DOI] [PubMed] [Google Scholar]
  • 117.Wang J, Zhao W, Guo Y, Zhang B, Xie Q, Xiang D, Gao J, Wang B, Chen Z. The expression of RNA-binding protein HuR in non-small cell lung cancer correlates with vascular endothelial growth factor-C expression and lymph node metastasis. Oncology. 2009;76:420–429. doi: 10.1159/000216837. [DOI] [PubMed] [Google Scholar]
  • 118.Evans JF, Kargman SL. Cancer and cyclooxygenase-2 (COX-2) inhibition. Curr Pharm Des. 2004;10:627–634. doi: 10.2174/1381612043453126. [DOI] [PubMed] [Google Scholar]
  • 119.Sahin M, Sahin E, Gümüslü S. Cyclooxygenase-2 in cancer and angiogenesis. Angiology. 2009;60:242–253. doi: 10.1177/0003319708318378. [DOI] [PubMed] [Google Scholar]
  • 120.Lanas A. Nonsteroidal antiinflammatory drugs and cyclooxygenase inhibition in the gastrointestinal tract: a trip from peptic ulcer to colon cancer. Am J Med Sci. 2009;338:96–106. doi: 10.1097/MAJ.0b013e3181ad8cd3. [DOI] [PubMed] [Google Scholar]
  • 121.Dixon DA, Tolley ND, King PH, Nabors LB, McIntyre TM, Zimmerman GA, Prescott SM. Altered expression of the mRNA stability factor HuR promotes cyclooxygenase-2 expression in colon cancer cells. J Clin Invest. 2001;108:1657–1665. doi: 10.1172/JCI12973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Young LE, Sanduja S, Bemis-Standoli K, Pena EA, Price RL, Dixon DA. The mRNA binding proteins HuR and tristetraprolin regulate cyclooxygenase 2 expression during colon carcinogenesis. Gastroenterology. 2009;136:1669–1679. doi: 10.1053/j.gastro.2009.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lim SJ, Lee SH, Joo SH, Song JY, Choi SI. Cytoplasmic Expression of HuR is Related to Cyclooxygenase-2 Expression in Colon Cancer. Cancer Res Treat. 2009;41:87–92. doi: 10.4143/crt.2009.41.2.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Barbisan F, Mazzucchelli R, Santinelli A, López-Beltran A, Cheng L, Scarpelli M, Montorsi F, Montironi R. Overexpression of ELAV-like Protein HuR is Associated with Increased COX-2 Expression in Atrophy, High-grade Prostatic Intraepithelial Neoplasia, and Incidental Prostate Cancer in Cystoprostatectomies. Eur Urol. 2008 doi: 10.1016/j.eururo.2008.04.043. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 125.Erkinheimo TL, Lassus H, Sivula A, Sengupta S, Furneaux H, Hla T, Haglund C, Butzow R, Ristimaki A. Cytoplasmic HuR expression correlates with poor outcome and with cyclooxygenase 2 expression in serous ovarian carcinoma. Cancer Res. 2003;63:7591–7594. [PubMed] [Google Scholar]
  • 126.Stoppoloni D, Cardillo I, Verdina A, Vincenzi B, Menegozzo S, Santini M, Sacchi A, Baldi A, Galati R. Expression of the embryonic lethal abnormal vision-like protein HuR in human mesothelioma: association with cyclooxygenase-2 and prognosis. Cancer. 2008;113:2761–2769. doi: 10.1002/cncr.23904. [DOI] [PubMed] [Google Scholar]
  • 127.Watnick RS, Cheng YN, Rangarajan A, Ince TA, Weinberg RA. Ras modulates Myc activity to repress thrombospondin-1 expression and increase tumor angiogenesis. Cancer Cell. 2003;3:219–231. doi: 10.1016/s1535-6108(03)00030-8. [DOI] [PubMed] [Google Scholar]
  • 128.Zabrenetzky V, Harris CC, Steeg PS, Roberts DD. Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines. Int J Cancer. 1994;59:191–195. doi: 10.1002/ijc.2910590209. [DOI] [PubMed] [Google Scholar]
  • 129.Keyse SM. Dual-specificity MAP kinase phosphatases (MKPs) and cancer. Cancer Metastasis Rev. 2008;27:253–261. doi: 10.1007/s10555-008-9123-1. [DOI] [PubMed] [Google Scholar]
  • 130.Wang X, Liu Y. Regulation of innate immune response by MAP kinase phosphatase-1. Cell Signal. 2007;19:1372–1382. doi: 10.1016/j.cellsig.2007.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Elliott RL, Blobe GC. Role of transforming growth factor Beta in human cancer. J Clin Oncol. 2005;23:2078–93. doi: 10.1200/JCO.2005.02.047. [DOI] [PubMed] [Google Scholar]
  • 132.Bierie B, Moses HL. Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer. 2006;6:506–520. doi: 10.1038/nrc1926. [DOI] [PubMed] [Google Scholar]
  • 133.Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001;29:117–129. doi: 10.1038/ng1001-117. [DOI] [PubMed] [Google Scholar]
  • 134.Beck C, Schreiber H, Rowley D. Role of TGF-beta in immune-evasion of cancer. Microsc Res Tech. 2001;52:387–395. doi: 10.1002/1097-0029(20010215)52:4<387::AID-JEMT1023>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
  • 135.Torre-Amione G, Beauchamp RD, Koeppen H, Park BH, Schreiber H, Moses HL, Rowley DA. A highly immunogenic tumor transfected with a murine transforming growth factor type beta 1 cDNA escapes immune surveillance. Proc Natl Acad Sci USA. 1990;87:1486–1490. doi: 10.1073/pnas.87.4.1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Nabors LB, Gillespie GY, Harkins L, King PH. HuR, a RNA stability factor, is expressed in malignant brain tumors and binds to adenine- and uridine-rich elements within the 3′ untranslated regions of cytokine and angiogenic factor mRNAs. Cancer Res. 2001;61:2154–2161. [PubMed] [Google Scholar]
  • 137.Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer. 2007;7:415–428. doi: 10.1038/nrc2131. [DOI] [PubMed] [Google Scholar]
  • 138.Dong R, Lu JG, Wang Q, He XL, Chu YK, Ma QJ. Stabilization of Snail by HuR in the process of hydrogen peroxide induced cell migration. Biochem Biophys Res Commun. 2007;356:318–321. doi: 10.1016/j.bbrc.2007.02.145. [DOI] [PubMed] [Google Scholar]
  • 139.Kerrigan JJ, Mansell JP, Sandy JR. Matrix turnover. J Orthod. 2000;27:227–233. doi: 10.1179/ortho.27.3.227. [DOI] [PubMed] [Google Scholar]
  • 140.Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2000;2:161–174. doi: 10.1038/nrc745. [DOI] [PubMed] [Google Scholar]
  • 141.Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25:9–34. doi: 10.1007/s10555-006-7886-9. [DOI] [PubMed] [Google Scholar]
  • 142.Libra M, Scalisi A, Vella N, Clementi S, Sorio R, Stivala F, Spandidos DA, Mazzarino C. Uterine cervical carcinoma: role of matrix metalloproteinases. Int J Oncol. 2009;34:897–903. doi: 10.3892/ijo_00000215. [DOI] [PubMed] [Google Scholar]
  • 143.Takeuchi T, Hisanaga M, Nagao M, Ikeda N, Fujii H, Koyama F, Mukogawa T, Matsumoto H, Kondo S, Takahashi C, Noda M, Nakajima Y. The membrane-anchored matrix metalloproteinase (MMP) regulator RECK in combination with MMP-9 serves as an informative prognostic indicator for colorectal cancer. Clin Cancer Res. 2004;10:5572–5579. doi: 10.1158/1078-0432.CCR-03-0656. [DOI] [PubMed] [Google Scholar]
  • 144.Clark JC, Thomas DM, Choong PF, Dass CR. RECK--a newly discovered inhibitor of metastasis with prognostic significance in multiple forms of cancer. Cancer Metastasis Rev. 2007;26:675–683. doi: 10.1007/s10555-007-9093-8. [DOI] [PubMed] [Google Scholar]
  • 145.Agarwal R, Agarwal C, Ichikawa H, Singh RP, Aggarwal BB. Anticancer potential of silymarin: from bench to bed side. Anticancer Res. 2006;26:4457–4498. [PubMed] [Google Scholar]
  • 146.Akool el S, Kleinert H, Hamada FM, Abdelwahab MH, Forstermann U, Pfeilschifter J, Eberhardt W. Nitric oxide increases the decay of matrix metalloproteinase 9 mRNA by inhibiting the expression of mRNA-stabilizing factor HuR. Mol Cell Biol. 2003;23:4901–4916. doi: 10.1128/MCB.23.14.4901-4916.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Huwiler A, Akool el S, Aschrafi A, Hamada FM, Pfeilschifter J, Eberhardt W. ATP potentiates interleukin-1 beta-induced MMP-9 expression in mesangial cells via recruitment of the ELAV protein HuR. J Biol Chem. 2003;278:51758–69. doi: 10.1074/jbc.M305722200. [DOI] [PubMed] [Google Scholar]
  • 148.Sidenius N, Blasi F. The urokinase plasminogen activator system in cancer: recent advances and implication for prognosis and therapy. Cancer Metastasis Rev. 2003;22:205–222. doi: 10.1023/a:1023099415940. [DOI] [PubMed] [Google Scholar]
  • 149.Mazar AP. Urokinase plasminogen activator receptor choreographs multiple ligand interactions: implications for tumor progression and therapy. Clin Cancer Res. 2008;14:5649–5655. doi: 10.1158/1078-0432.CCR-07-4863. [DOI] [PubMed] [Google Scholar]
  • 150.D’Alessio S, Blasi F. The urokinase receptor as an entertainer of signal transduction. Front Biosci. 2009;14:4575–4587. doi: 10.2741/3550. [DOI] [PubMed] [Google Scholar]
  • 151.Heinonen M, Bono P, Narko K, Chang SH, Lundin J, Joensuu H, Furneaux H, Hla T, Haglund C, Ristimaki A. Cytoplasmic HuR expression is a prognostic factor in invasive ductal breast carcinoma. Cancer Res. 2005;65:2157–2161. doi: 10.1158/0008-5472.CAN-04-3765. [DOI] [PubMed] [Google Scholar]
  • 152.Suswam EA, Nabors LB, Huang Y, Yang X, King PH. IL-1beta induces stabilization of IL-8 mRNA in malignant breast cancer cells via the 3′ untranslated region: Involvement of divergent RNA-binding factors HuR, KSRP and TIAR. Int J Cancer. 2005;113:911–919. doi: 10.1002/ijc.20675. [DOI] [PubMed] [Google Scholar]
  • 153.Pryzbylkowski P, Obajimi O, Keen JC. Trichostatin A and 5 Aza-2′ deoxycytidine decrease estrogen receptor mRNA stability in ER positive MCF7 cells through modulation of HuR. Breast Cancer Res Treat. 2008;111:15–25. doi: 10.1007/s10549-007-9751-0. [DOI] [PubMed] [Google Scholar]
  • 154.Woo HH, Zhou Y, Yi X, David CL, Zheng W, Gilmore-Hebert M, Kluger HM, Ulukus EC, Baker T, Stoffer JB, Chambers SK. Regulation of non-AU-rich element containing c-fms proto-oncogene expression by HuR in breast cancer. Oncogene. 2009;28:1176–1186. doi: 10.1038/onc.2008.469. [DOI] [PubMed] [Google Scholar]
  • 155.Hostetter C, Licata LA, Witkiewicz A, Costantino CL, Yeo CJ, Brody JR, Keen JC. Cytoplasmic accumulation of the RNA binding protein HuR is central to tamoxifen resistance in estrogen receptor positive breast cancer cells. Cancer Biol Ther. 2008;7:1496–506. doi: 10.4161/cbt.7.9.6490. [DOI] [PubMed] [Google Scholar]
  • 156.Saunus JM, French JD, Edwards SL, Beveridge DJ, Hatchell EC, Wagner SA, Stein SR, Davidson A, Simpson KJ, Francis GD, Leedman PJ, Brown MA. Posttranscriptional regulation of the breast cancer susceptibility gene BRCA1 by the RNA binding protein HuR. Cancer Res. 2008;68:9469–9478. doi: 10.1158/0008-5472.CAN-08-1159. [DOI] [PubMed] [Google Scholar]
  • 157.Heinonen M, Fagerholm R, Aaltonen K, Kilpivaara O, Aittomäki K, Blomqvist C, Heikkilä P, Haglund C, Nevanlinna H, Ristimäki A. Prognostic role of HuR in hereditary breast cancer. Clin Cancer Res. 2007;13:6959–6963. doi: 10.1158/1078-0432.CCR-07-1432. [DOI] [PubMed] [Google Scholar]
  • 158.Denkert C, Koch I, von Keyserlingk N, Noske A, Niesporek S, Dietel M, Weichert W. Expression of the ELAV-like protein HuR in human colon cancer: association with tumor stage and cyclooxygenase-2. Mod Pathol. 2006;19:1261–1269. doi: 10.1038/modpathol.3800645. [DOI] [PubMed] [Google Scholar]
  • 159.Lu Z, Luo RZ, Peng H, Rosen DG, Atkinson EN, Warneke C, Huang M, Nishmoto A, Liu J, Liao WS, Yu Y, Bast RC., Jr Transcriptional and posttranscriptional down-regulation of the imprinted tumor suppressor gene ARHI (DRAS3) in ovarian cancer. Clin Cancer Res. 2006;12:2404–2413. doi: 10.1158/1078-0432.CCR-05-1036. [DOI] [PubMed] [Google Scholar]
  • 160.Denkert C, Weichert W, Pest S, Koch I, Licht D, Kobel M, Reles A, Sehouli J, Dietel M, Hauptmann S. Overexpression of the embryonic-lethal abnormal vision-like protein HuR in ovarian carcinoma is a prognostic factor and is associated with increased cyclooxygenase 2 expression. Cancer Res. 2004;64:189–195. doi: 10.1158/0008-5472.can-03-1987. [DOI] [PubMed] [Google Scholar]
  • 161.Yi X, Zhou Y, Zheng W, Chambers SK. HuR expression in the nucleus correlates with high histological grade and poor disease-free survival in ovarian cancer. Aust N Z J Obstet Gynaecol. 2009;49:93–98. doi: 10.1111/j.1479-828X.2008.00937.x. [DOI] [PubMed] [Google Scholar]
  • 162.Niesporek S, Kristiansen G, Thoma A, Weichert W, Noske A, Buckendahl AC, Jung K, Stephan C, Dietel M, Denkert C. Expression of the ELAV-like protein HuR in human prostate carcinoma is an indicator of disease relapse and linked to COX-2 expression. Int J Oncol. 2008;32:341–347. [PubMed] [Google Scholar]
  • 163.Costantino CL, Witkiewicz AK, Kuwano Y, Cozzitorto JA, Kennedy EP, Dasgupta A, Keen JC, Yeo CJ, Gorospe M, Brody JR. The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR Up-regulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase. Cancer Res. 2009;69:4567–4572. doi: 10.1158/0008-5472.CAN-09-0371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Cho NP, Han HS, Soh Y, Son HJ. Overexpression of cyclooxygenase-2 correlates with cytoplasmic HuR expression in salivary mucoepidermoid carcinoma but not in pleomorphic adenoma. J Oral Pathol Med. 2007;36:297–303. doi: 10.1111/j.1600-0714.2007.00526.x. [DOI] [PubMed] [Google Scholar]
  • 165.Hasegawa H, Kakuguchi W, Kuroshima T, Kitamura T, Tanaka S, Kitagawa Y, Totsuka Y, Shindoh M, Higashino F. HuR is exported to the cytoplasm in oral cancer cells in a different manner from that of normal cells. Br J Cancer. 2009;100:1943–1948. doi: 10.1038/sj.bjc.6605084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Mrena J, Wiksten JP, Thiel A, Kokkola A, Pohjola L, Lundin J, Nordling S, Ristimäki A, Haglund C. Cyclooxygenase-2 is an independent prognostic factor in gastric cancer and its expression is regulated by the messenger RNA stability factor HuR. Clin Cancer Res. 2005;11:7362–7368. doi: 10.1158/1078-0432.CCR-05-0764. [DOI] [PubMed] [Google Scholar]
  • 167.Mrena J, Wiksten JP, Kokkola A, Nordling S, Haglund C, Ristimaki A. Prognostic significance of cyclin A in gastric cancer. Int J Cancer. 2006;119:1897–1901. doi: 10.1002/ijc.21944. [DOI] [PubMed] [Google Scholar]
  • 168.Koljonen V, Böhling T, Haglund C, Ristimäki A. Expression of HuR in Merkel cell carcinoma and in normal skin. J Cutan Pathol. 2008;35:10–14. doi: 10.1111/j.1600-0560.2007.00762.x. [DOI] [PubMed] [Google Scholar]
  • 169.Abdelmohsen K, Srikantan S, Kuwano Y, Gorospe M. miR-519 reduces cell proliferation by lowering RNA-binding protein HuR levels. Proc Natl Acad Sci USA. 2008;105:20297–20302. doi: 10.1073/pnas.0809376106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Guo X, Wu Y, Hartley RS. MicroRNA-125a represses cell growth by targeting HuR in breast cancer. RNA Biol. 2009;6 doi: 10.4161/rna.6.5.10079. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Meisner NC, Hintersteiner M, Mueller K, Bauer R, Seifert JM, Naegeli HU, Ottl J, Oberer L, Guenat C, Moss S, Harrer N, Woisetschlaeger M, Buehler C, Uhl V, Auer M. Identification and mechanistic characterization of low-molecular-weight inhibitors for HuR. Nat Chem Biol. 2007;3:508–515. doi: 10.1038/nchembio.2007.14. [DOI] [PubMed] [Google Scholar]
  • 172.Ortega AD, Sala S, Espinosa E, González-Barón M, Cuezva JM. HuR and the bioenergetic signature of breast cancer: a low tumor expression of the RNA-binding protein predicts a higher risk of disease recurrence. Carcinogenesis. 2008;29:2053–2061. doi: 10.1093/carcin/bgn185. [DOI] [PubMed] [Google Scholar]
  • 173.López de Silanes I, Fan J, Galbán CJ, Spencer RG, Becker KG, Gorospe M. Global analysis of HuR-regulated gene expression in colon cancer systems of reducing complexity. Gene Expr. 2004;12:49–59. doi: 10.3727/000000004783992215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Papadaki O, Milatos S, Grammenoudi S, Mukherjee N, Keene JD, Kontoyiannis DL. Control of thymic T cell maturation, deletion and egress by the RNA-binding protein HuR. J Immunol. 2009;182:6779–6788. doi: 10.4049/jimmunol.0900377. [DOI] [PubMed] [Google Scholar]

RESOURCES