Next Article in Journal
Systematic Review of Available CAR-T Cell Trials around the World
Next Article in Special Issue
Establishment and Characterization of a New Human Intrahepatic Cholangiocarcinoma Cell Line LIV27
Previous Article in Journal
Ficus dubia Latex Extract Induces Cell Cycle Arrest and Apoptosis by Regulating the NF-κB Pathway in Inflammatory Human Colorectal Cancer Cell Lines
Previous Article in Special Issue
Potential Role of Inflammation-Promoting Biliary Microbiome in Primary Sclerosing Cholangitis and Cholangiocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hu Antigen R (HuR) Protein Structure, Function and Regulation in Hepatobiliary Tumors

by
Sofia Lachiondo-Ortega
1,
Teresa Cardoso Delgado
1,
Blanca Baños-Jaime
2,
Alejandro Velázquez-Cruz
2,
Irene Díaz-Moreno
2 and
María Luz Martínez-Chantar
1,3,*
1
Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
2
Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Instituto de Investigaciones Químicas (IIQ), Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), 41092 Sevilla, Spain
3
Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(11), 2666; https://doi.org/10.3390/cancers14112666
Submission received: 15 April 2022 / Revised: 19 May 2022 / Accepted: 23 May 2022 / Published: 27 May 2022
(This article belongs to the Special Issue Theranostic Advances in Hepatobiliary Tumors)

Abstract

:

Simple Summary

Hepatobiliary tumors are a group of primary malignancies encompassing the liver, the intra- and extra-hepatic biliary tracts, and the gall bladder. Within the liver, hepatocellular carcinoma (HCC) is the most common type of primary cancer, which is, also, representing the third-most recurrent cause of cancer-associated death and the sixth-most prevalent type of tumor worldwide, nowadays. Although less frequent, cholangiocarcinoma (CCA) is, currently, a fatal cancer with limited therapeutic options. Here, we review the regulatory role of Hu antigen R (HuR), a ubiquitous member of the ELAV/Hu family of RNA-binding proteins (RBPs), in the pathogenesis, progression, and treatment of HCC and CCA. Overall, HuR is proposed as a valuable diagnostic and prognostic marker, as well as a therapeutic target in hepatobiliary cancers. Therefore, novel therapeutic approaches that can selectively modulate HuR function appear to be highly attractive for the clinical management of these types of tumors.

Abstract

Hu antigen R (HuR) is a 36-kDa ubiquitous member of the ELAV/Hu family of RNA-binding proteins (RBPs), which plays an important role as a post-transcriptional regulator of specific RNAs under physiological and pathological conditions, including cancer. Herein, we review HuR protein structure, function, and its regulation, as well as its implications in the pathogenesis, progression, and treatment of hepatobiliary cancers. In particular, we focus on hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), tumors where the increased cytoplasmic localization of HuR and activity are proposed, as valuable diagnostic and prognostic markers. An overview of the main regulatory axes involving HuR, which are associated with cell proliferation, invasion, metastasis, apoptosis, and autophagy in HCC, is provided. These include the transcriptional, post-transcriptional, and post-translational modulators of HuR function, in addition to HuR target transcripts. Finally, whereas studies addressing the relevance of targeting HuR in CCA are limited, in the past few years, HuR has emerged as a potential therapeutic target in HCC. In fact, the therapeutic efficacy of some pharmacological inhibitors of HuR has been evaluated, in early experimental models of HCC. We, further, discuss the major findings and future perspectives of therapeutic approaches that specifically block HuR interactions, either with post-translational modifiers or cognate transcripts in hepatobiliary cancers.

Graphical Abstract

1. Introduction

Hu antigen R (HuR), also known as HuA or embryonic lethal abnormal vision-like protein 1 (ELAVL1), is a 36-kDa ubiquitous member of the ELAV/Hu family of RNA-binding proteins (RBPs), which, also, includes HuB, HuC and HuD [1,2]. In humans, this protein is encoded by the ELAVL1 gene. In healthy individuals, HuR protein is, mostly, expressed in human endocrine tissues, the respiratory system, and the gastrointestinal tract [3].

1.1. HuR Protein Structure

HuR structure is, mainly, comprised of three RNA-recognition motifs (RRMs) (Figure 1A) [4]. The first two domains (RRM1 and RRM2) are connected by a 10-residue linker in a tandem structure, as revealed by its X-ray crystallographic model (Figure 1B, middle panel), and are the main domains involved in RNA recognition [5]. Despite the high dynamics of the N-terminal unstructured 20-residue stretch of HuR, nuclear magnetic resonance (NMR) experimental restraints revealed that it was folded towards the RRM1 β-sheet (Figure 1B, left panel) [6]. Interestingly, it harbors the HuR redox sensor Cys13, which is conserved among ELAVL1 orthologous proteins and is involved in HuR RRM1 and RRM1-2 homodimerization and, to some extent, in the regulation of the antioxidant response by HuR (Figure 1C) [7,8]. A 60-residue disordered hinge region spaces out the C-terminal RRM3 domain from the tandem RRM1-2. This linker region contains the HuR nucleocytoplasmic shuttling sequence (HNS) (Figure 1A), whose length enables transient contacts between RRM3 and RRM1–2 [9]. Phosphorylation at HNS modifies HuR interaction with nucleo-cytoplasmic transport proteins, triggering HuR accumulation in the cytosol [10,11]. HuR RRM3, also, forms dimers through its evolutionary well-conserved Trp261 residue along evolution (Figure 1C), which is located in helix α1 of RRM3 (Figure 1B, right panel) [12,13].
All three RRMs show the canonical β1α1β2β3α2β4 topology, with a β-sheet formed by four antiparallel β-strands folded against two α-helices. The two central β-strands (β1 and β3) of each RRM contain the ribonucleoprotein (RNP) motifs RNP-1 (in β3) and RNP-2 (in β1), which are essential for RNA/DNA binding (Figure 1B,C) [15,16]. HuR RRM domains bind single-stranded RNA/DNA, by means of stacking interactions and hydrogen bonds between RNP motifs and nucleic acids [17]. RNP motif sequences are indicated in Figure 1C and, even though they differ for each RRM of HuR, they have been extensively conserved in ELAV-like proteins, through evolution from Drosophila to vertebrates [18].

1.2. HuR Protein Function

HuR functions as a post-transcriptional regulator, affecting many aspects of RNA metabolism, from splicing to translation, by binding through its RRMs to AU-rich elements (AREs), typically present in the 3′-untranslated region (UTR) of its target transcripts [19,20]. Under physiological conditions, HuR is, predominantly, located in the nucleus, where it participates in pre-mRNA splicing and nuclear export of mature mRNAs [21,22,23,24,25]. Notably, its nuclear functions remain poorly understood. Upon specific stimuli (such as stress signals and mitogens), HuR translocates to the cytoplasm and develops its main biological functions, which are mRNA stabilization and modulation of translation. HuR nucleo-cytoplasmic shuttling, through the nucleopore, is aided by the import factors transportin-1 (Trn1) and -2 (Trn2), and the adaptor proteins ANP32A (pp32/PHAP-I) and ANP32B (APRIL), both independently mediating HuR association with the nuclear export receptor chromosome region maintenance 1 (CRM1) [22,26,27].
In the cytoplasm, it is thought that HuR might control RNA stability, by competing with other RBPs and, thus, preventing mRNA degradation and deadenylation. To date, over 80 HuR-stabilized target mRNAs have been described and reviewed in [28,29], which include those that encode c-Fos [30], p21 [31,32,33], cyclins A2 [30,34], B1 [30,34], E1 [35,36], D1 [30,37], inducible nitric oxide synthase (iNOS) [38,39], granulocyte macrophage-colony stimulating factor (GM-CSF) [40,41], eukaryotic initiation factor (eIF)-4E [42], murine double minute (Mdm)2 [43,44], vascular endothelial growth factor (VEGF) [45,46,47,48,49,50], transforming growth factor (TGF)-β [45], sirtuin 1 (SIRT1) [51,52], tumor necrosis factor (TNF)-α [40,45,53], B-cell leukemia (Bcl)-2 [54,55], myeloid leukemia cell differentiation protein (Mcl)-1 [54], oncostatin M (OSM) [56], cyclooxygenase (COX)-2 [30,45,52,57,58,59,60,61,62], γ-glutamylcysteine synthetase heavy subunit (γ-GCSh) [63], survival of motor neuron (SMN) [64], SH2D1A [65], the regulator of G-protein signaling 4 (RGS4) [66], parathyroid hormone-related protein (PTHrP) [67], Fas ligand (FasL) [68], Myogenin [69,70], MyoD [69,70], acetylcholinesterase (AChE) [71,72], p53 [73,74], ARHI [aplasia Ras homolog member I (DIRAS3)] [58], nitric oxide/soluble guanylyl cyclase (sGC) [75], urokinase plasminogen activator (uPA) and its receptor (uPAR) [76], neurofibromatosis type 1 (NF1) [77], von Hippel-Lindau protein (pVHL) [67,78], toll-like receptor 4 (TLR4) [79], Snail [80], matrix metalloprotease (MMP)-9 [81,82,83,84,85], c-Fms [86], the MAPK phosphatase (MKP)-1 [87], interferon (IFN)-γ [88], interleukin (IL)-3 [89], IL-4 [90], IL-6 [45,91,92], IL-8 [45,57,93], and HuR itself [25,94,95].
Regarding the control of translation, HuR can promote the expression of many mRNAs that are templates for proteins, such as cyclin A2 [30,34,96], prothymosin α (ProTα) [97], hypoxia-inducible factor (HIF)-1α [98], Bcl-2 [54,55,99], VEGF [45,46,47,49,100,101,102], thrombospondin (TSP)-1 [103], MKP-1 [87], p53 [73,74], the cationic amino acid transporter (CAT)-1 [104], the intrinsic cellular caspase inhibitor XIAP [37], and cytochrome c [105]. Importantly, HuR can also bind to the 5′-UTR of a small subset of target transcripts (e.g., p27, IGF-1R and thrombomodulin) and disrupt the internal ribosome entry site (IRES)-dependent translation, eventually resulting in the repression of protein expression [106,107,108]. HuR was, also, found to bind to the 3′-UTR of Wnt5a and c-Myc mRNAs and repress their translation [109,110].
In addition to coding transcripts, HuR has the ability to bind and regulate the function of microRNAs (miRNAs) [111,112,113,114] and long non-coding RNAs (lncRNAs) [111,115,116,117].

1.3. Regulation of HuR Protein Function

The function of HuR is regulated through transcriptional, post-transcriptional, and post-translational mechanisms, as discussed below.

1.3.1. Transcriptional Control of HuR

The ELAVL1 gene displays alternate transcriptional start sites, leading to at least two distinct transcripts with distinct 5′-UTRs [99,118], the shorter of them being expressed in stress conditions under the influence of bone morphogenetic protein-7 (BMP-7) and the Smad 1/5/8 route [118]. Expression of the long HuR isoform relies on nuclear factor k of activated B-cells (NF-kB). The HuR promoter region contains a target of NF-kB, to which one of its members—p65/RelA—binds upon the induction of the phosphatidylinositol 3-kinase (PI3K)/AKT signaling route, resulting in the abnormal upregulation of HuR gene expression in gastric tumors [119].

1.3.2. Post-Transcriptional Control of HuR

The stability of HuR mRNA is another key factor controlling its protein levels. HuR can both up- and down-regulate its expression, by interacting with its own transcript [25,94,95,120]. In addition, the existence of both positive and negative feedback loops between HuR and the protein product of its target mRNAs have been reported [121,122,123,124].
HuR abundance can, also, be regulated by miRNAs. An assortment of miRNAs (miR-125 [125], miR-519 [126], miR-9 [127], 291b-3p [128], miR-570-3 [129], miR-16 [130], and miR-22 [131]) has been found to target the coding region and 3′-UTR of HuR mRNA, suppressing its expression. Conversely, HuR 3′-UTR comprises a binding site for miRNA-155-5p, which stabilizes the transcript and favors HT-29 colorectal cancer (CRC) cell migration [132].

1.3.3. Post-Translational Control of HuR

Post-translational modifications (PTMs) account for the main mechanism of regulation for HuR function, allowing the protein to elicit quick changes in gene-expression programs [10,11]. HuR protein abundance, subcellular localization, and RNA-binding affinity can be modulated by methylation [133,134,135,136], phosphorylation [24,33,44,51,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157], proteolytic cleavage [158,159,160], ubiquitination [78,147,161,162], PARylation [163,164], sulfhydration [165], and arginylation [166].
More recently, it was described that circular RNAs (circRNAs) may comprise various binding motifs for one or several RBPs, including HuR, acting as sponges, where the RBP is prevented from binding to other targets while tethered to the circRNA [167]. For instance, it was shown that circPABPN1 competes with PABP1 mRNA for binding to HuR, affecting the expression of p53, c-myc, and bcl-2, besides PABP1 [168].

1.3.4. HuR Protein Oligomerization

HuR and functionally related proteins, such as AUF1, form dimers and multimers both free and bound to target sequences [12,15,153,169,170,171]. HuR dimers have been detected by fluorescence resonance energy transfer (FRET), both in the cell nucleus and cytoplasm. Importantly, the HuR oligomerization state strongly affects the stability of its target mRNAs. Fluorescence anisotropy analyses suggested that HuR RRM1-2 is able to form dimers with high affinity (KD ca. 0.1 nM). These data indicate that RRM1-2 dimerization could be an important feature of the interaction mechanism between HuR and its target RNAs [172]. Furthermore, a structural analysis revealed that RRM1 alone forms homodimers as well, since Cys13 is exposed to and capable of establishing disulfide bonds, thus enabling HuR to respond to oxidative stress [7]. Beyond dimerization, cytoplasmic multimerization of HuR has been observed in glioma cells. A thorough analysis using distinct HuR constructs bound to fluorescent and bioluminescent probes enabled a model to be constructed, in which Cys13 is responsible for covalent dimerization, although the hinge region and RRM3 are still necessary for further oligomerization [173]. Indeed, the C-terminal domain of HuR is, also, prominently involved in its dimerization [12,13,15,170], as well as in the cooperative formation of HuR oligomers on ARE motifs [12,153,170]. According to small-angle X-ray scattering (SAXS) and NMR data, a single site-directed mutation (W261E) in RRM3 was enough to hinder its dimerization, under reducing conditions [15]. Moreover, deletion of RRM3 critically impaired the ability of the RBP to stabilize reporter β-globin mRNA constructs containing different AREs [9]. Altogether, experimental evidence points to HuR dimerizing through RRM1 and RRM3 in an independent way, while requiring both domains for oligomerization.

2. HuR and Hepatobiliary Cancers

HuR owns the post-transcriptional control of a large number of RNAs, enabling the protein to play pivotal roles that are dictated by the molecular functions of the transcripts. HuR targets, chiefly, include many mRNAs encoding proteins involved in cell proliferation, senescence, apoptosis, differentiation, stress, and immune responses. In turn, HuR was found to be involved not only in physiological processes (e.g., adipogenesis and muscle differentiation) [69,174,175,176] but also in disease (primarily, cancer and inflammation) [28,29,177]. Indeed, the relevance of HuR in some types of hepatobiliary tumors has been constantly described during the last decade. The major findings to date and future perspectives on the applications of HuR as a protein marker for the diagnosis, prognosis, and therapy of hepatobiliary cancers are the main topic of this review.

2.1. Hepatobiliary Cancers

Hepatobiliary cancers are a group of primary malignancies encompassing the liver, the intra- and extra-hepatic biliary tracts, and the gall bladder. Within the liver, the most common primary cancer is hepatocellular carcinoma (HCC). HCC, usually, arises in patients with an underlying chronic liver disease, such as chronic infection with the hepatitis B (HBV) or C virus (HBC), cirrhosis, excessive alcohol consumption, metabolic syndrome, diabetes, and non-alcoholic fatty liver disease (NAFLD) [178]. Even though the clinical management of HCC has improved in the past ten years, particularly for patients at advanced stages, HCC is still the third-most common cause of cancer-associated death and the sixth-most prevalent type of cancer worldwide [179]. Of relevance, in the past few years, important studies on immunotherapy in HCC have been carried out, resulting in novel guidelines for its treatment [180,181,182].
On the other hand, cholangiocarcinoma (CCA) encompasses a group of malignancies, arising at any point in the hepatic biliary tree. Biliary tract cancers, including intrahepatic (ICC), perihilar (PCC), and distal cholangiocarcinoma (DCC), as well as gallbladder cancer, are low-incidence malignancies in most high-income countries, but represent a major health problem in endemic areas. Indeed, ICC is the second-leading cause of primary liver cancer [183]. Nowadays, CCA is a fatal cancer, with a survival rate beyond a year of diagnosis inferior to 5% [184]. Therapeutic options are limited and surgery is the cornerstone of cure for CCA, even though most patients present with locally advanced or metastatic disease [185]. In the past few years, druggable alterations such as fibroblast growth factor receptor 2 (FGFR2) gene fusions and rearrangements, or isocitrate dehydrogenase-1 (IDH-1) and BRAF mutations, have been widely described in CCA patients, further indicating the important differences between iCCA and PCC/DCC, as thoroughly reviewed in [186].

2.2. HuR as a Prognostic and Diagnostic Biomarker in Hepatobiliary Cancers

HuR protein is, either, overexpressed in most human cancers or overactivated, as denoted by its increased cytoplasmic localization and the translation of various mRNAs involved in carcinogenesis. Regarding liver cancer, Embade et al. [187], reported significantly higher HuR protein levels in the mouse liver progenitor 29 (MLP29) cell line and in the S-adenosylmethionine-deficient (SAMe-D) cell line, isolated from the methionine adenosyltransferase (MAT)1A knockout (MAT1A-KO) mouse model of HCC, compared with primary mouse hepatocytes. Accordingly, higher HuR expression levels were found in human hepatoma cells (e.g., HepG2, Hep3B, SNU398, SNU449, SNU182, and SNU475), than in normal CRL4020 cells [188]. Moreover, in a human liver cancer tissue microarray (TMA) of 59 liver tissue cores from 44 patients, also by Zhu et al. [188], HCC tumor tissues showed significantly higher overall and cytoplasmic HuR staining, compared to normal liver tissues, and this high HuR staining score correlated with worse survival of patients with early-stage HCC. Furthermore, immunofluorescence analyses of normal versus malignant liver tissue revealed that HuR protein is down-regulated in normal human liver samples and up-regulated in HCC samples of different aetiologies (cirrhotic patients with HCV, alcoholic steatohepatitis, and non-alcoholic steatohepatitis (NASH)), where HuR concentration increased, proportionately, to their transformation status [189]. Finally, the expression of ELAVL1 gene is highly induced in the tumor tissue of a cohort of CCA patients, according to The Cancer Genome Atlas (TCGA) mRNA expression repository [190]. Likewise, high cytoplasmic HuR levels are associated with poor survival in patients with surgically resected CCA, treated with adjuvant gemcitabine-based chemotherapy [191].
In sum, HuR overexpression, along with its cytoplasmic localization, are hallmarks of both HCC and CCA, correlating with disease progression and overall survival. Even though the role of HuR in HCC has been widely investigated, with some studies revealing HuR targets and regulators under these conditions, the function of HuR in CCA remains rather unexplored to date.

2.3. Signaling Pathways Implicated in HCC Involving HuR

The oncogenic-gene-expression programs that allow cancer cells to develop, survive, proliferate, and colonize other tissues are strongly dependent on post-transcriptional mechanisms [28]. As has been previously described, HuR is highly involved in many types of cancer, including HCC and CCA, and so are the numerous HuR-regulated RNAs, which are known to contribute to the main cancer hallmark functions (i.e., enhanced cell proliferation and survival, elevated local angiogenesis, evasion of immune recognition, facilitated cancer cell invasion, and metastasis). Furthermore, a series of transcriptional, post-transcriptional, and post-translational regulators of HuR function have been found to be altered during HCC. Herein, we aimed to analyze the main regulatory axes involving HuR that are associated with hepatobiliary tumors, which can be classified into those related with cell proliferation, invasion, and metastasis; apoptosis; and autophagy (Figure 2). In the process, some of the previously reported HuR RNA targets and regulators of HuR function have been confirmed in the specific context of HCC, while others have been newly identified in this type of tumor.

2.3.1. The Role of HuR in Cell Proliferation, Migration, and Metastasis during HCC

Pathways Involving Coding Transcripts

The PI3K/AKT signaling route is known to be upregulated in HCC [192], and, therefore, it could be responsible for the increased expression of the long HuR mRNA isoform, through NF-kB, as it occurs in gastric tumors [119]. However, it is likely that expression of the short HuR transcript is not observed during HCC, as reduced levels of BMP-7 and p-Smad1/5/8 were detected in patient samples [193].
During HBV-associated HCC, the HBV-encoded X (HBx) protein upregulates HuR expression, which enhances HER2 mRNA stabilization and translation, thus contributing to the migration of HCC cells [194]. In the MAT1A knockout (MAT1A-KO) mouse model, which shows a chronic deficiency in SAMe levels and, spontaneously, develops NASH and HCC, hepatic levels of LKB1 and AMPK are activated, incrementing the cytoplasmic localization of HuR, which leads to the stabilization and expression of cyclin A2 and D1 mRNAs, and subsequent cell cycle progression [195]. Moreover, an abnormally low ratio between methylated and unmethylated HuR was revealed in HCC samples. The two HuR isoforms can associate with the 3′-UTR of MAT2A mRNA, whose activity is linked to liver cell proliferation. However, while unmethylated HuR was shown to stabilize the MAT2A transcript, an increase in its methylation status correlated with lower MAT2A mRNA levels. Hence, the loss of HuR methylation may explain the increased MAT2A mRNA and protein expression, and the subsequent loss of SAMe homeostasis that occurs during hepatocyte dedifferentiation, proliferation, and carcinogenesis [189]. Moreover, related with PTMs, NEDDylation of HuR was firstly reported in the context of liver cancer. Specifically, the E3 ligase Mdm2 catalyzes the conjugation of NEDD8 to HuR at Lys283, Lys313, and Lys326, a process that has been linked to the nuclear localization and reduced proteasomal degradation of the RBP [187]. Interestingly, the Mdm2 transcript being a described target of HuR [43,44], and considering the significantly positive correlation between Mdm2 and HuR expression in clinical HCC and human hepatoma cell lines [187], it would be highly expected that the mRNA levels of this E3 ligase were stabilized by HuR during HCC too, despite not having been verified to date.
The Wnt/β-catenin pathway has been shown to induce stearoyl-CoA desaturase (SCD) expression in liver-tumor-initiating and HCC cells, further increasing the synthesis of mono unsaturated fatty acids (MUFAs). MUFAs can block the nuclear import of HuR, thereby increasing its protein levels in the cytoplasm, where it binds to the 3′UTR of Lrp5 and Lrp6 mRNAs as well as stabilizes and stimulates their translation, further providing a positive feedback loop, by amplifying Wnt/β-catenin signaling and contributing to liver carcinogenesis [196]. Moreover, Wilms tumor 1-associated protein (WTAP) drives N6-methyladenosine (m6A) RNA methylation and epigenetic silencing of ETS1, by interfering with HuR-mediated stabilization of ETS1 mRNA, further alleviating the expression of p21 and p27 G2/M checkpoint proteins, which are known downstream effectors of ETS1, and facilitating HCC progression [197]. Moreover, it has been reported that ionizing radiation activates the DNA damage response (DDR) via ATM/p38, which causes HuR shuttling to the cytoplasm in order to stabilize mitochondrial transcription factor A (TFAM) mRNA and induce its expression in HepG2 hepatoma cells. These results suggest a new pathway, which could be targeted to increase the sensitivity of liver cancer cells to radiotherapy [198].

Pathways Involving Non-Coding Transcripts

In addition to mRNAs, a few examples, whereby HuR interacts with non-coding transcripts during hepatobiliary tumors, have been reported. In human HCC cells subjected to hypoxic stress, HuR binds to the primary transcript of miR-199a (pri-miR-199a) blocking its processing into mature miR-199a. Interestingly, miR-199a is a negative regulator of Hk2 and Pkm2 mRNA expression. Therefore, HuR-mediated miR-199a maturation inhibition, during hypoxia, enables the metabolic reprogramming of HCC cells towards the Warburg effect, which confers favorable conditions for tumor growth, invasion, and metastasis [199]. The long intergenic noncoding RNA (lincRNA)-UFC1 plays an oncogenic role in liver cancer, by interacting with HuR, which stabilizes and induces the expression of the CTNNB1 mRNA, leading to increased cell-cycle progression as well as proliferation and reduced apoptosis in HCC cells [200]. The oncofetal lncRNA Ptn-dt appeared to be highly expressed in HCC tissue and was found to interact with HuR protein, further compromising the stabilization and expression of miR-96. Therefore, the reduced function of miR-96 on the post-transcriptional inhibition of anaplastic lymphoma kinase (Alk) protein contributed to HCC cell proliferation [201]. Another study, describing the tumor suppressor role of lncRNA-AK058003 in HCC, revealed SNCG mRNA as a potential target of HuR. It was postulated that lncRNA-AK058003 is downregulated during HCC but, if overexpressed, it can interact with HuR to suppresses its expression, further affecting SNCG translation and stability, thus inhibiting γ-synuclein-mediated HCC cell proliferation and metastasis, both in vitro and in vivo [202].
In an attempt to elucidate the function of circRNAs, a few circRNA-RBP-mRNA axes involving HuR were revealed in HCC. For example, circBACH1 acts as an oncogene during hepatic tumorigenesis, upon association with HuR, to facilitate its translocation to the cytoplasm, where the RBP inhibits p27 protein expression and allows cell cycle progression, eventually favoring HCC cell proliferation [203]. KIAA1429, a key component of the m6A methyltransferase complex, negatively regulates circRNA-DLC1 in HCC tissues. A mechanistic study revealed that circDLC1 competitively binds with HuR, thereby impairing HuR-mediated MMP1 mRNA stabilization and expression, ultimately resulting in decreased hepatoma cell proliferation and metastasis [204]. Moreover, hsa_circ_0074854 physically interacts and stabilizes HuR protein in the cytoplasm, which induces ZEB1 protein expression, thereby promoting the migration, invasion, and epithelial-mesenchymal transition (EMT) of HepG2 hepatoma cells [205].

2.3.2. The Role of HuR in Cell Death during HCC

In SAMe-D cells derived from MAT1A KO mice, sustained LKB1 phosphorylation contributes to increased cytoplasmic HuR localization, where it, specifically, binds to the 3′-UTR of herpesvirus-associated ubiquitin-specific protease (HAUSP) mRNA, stabilizing it and increasing its transcription. The subsequent accumulation of HAUSP deubiquitinating enzyme in the cytoplasm allows its interaction with p53, which increases the stability of the tumor suppressor in the cytoplasm, thereby controlling the apoptotic response [206].
A recent study about a possible model, whereby alpha fetoprotein (AFP) regulates HCC progression and chemosensitivity, reported the reactivation of AFP during hepatocarcinogenesis and its interaction with HuR, resulting in the redistribution of the RBP to the cytoplasm. There, HuR would bind to the 3′-UTR of the Fas death receptor mRNA and repress its translation, without affecting its stability or splicing, which further suppresses the Fas/FADD-mediated extrinsic apoptotic program and bypasses immune surveillance in HCC-derived cell lines [188,207].

2.3.3. The Role of HuR in Autophagy during HCC

Interestingly, HuR binds to the 3′-UTR of ATG5, ATG12, and ATG16 mRNAs and enhances their translation. As a result, autophagosome formation is enhanced, dysregulating the autophagy activity in HCC cell lines, which might possibly act as a pro-survival response and promote hepatic tumor growth [208]. On the other hand, inhibition of autophagy by BECN1 siRNA leads to HuR-enhanced ferroptosis in HCC. HuR and BECN1 interaction induces autophagosome formation, increasing autophagic ferritin degradation and enhancing ferroptosis in hepatic stellate cells (HSCs) [209].

3. Therapeutic Approaches to Inhibit HuR in Hepatobiliary Tumors

Given its essential role as a regulation hub of cell-fate decisions, suppression of HuR activity has become a key objective to control tumor progression and therapy resistance. Even though RBPs were, initially, considered unsuitable for drug screening, developments during the last decade have enabled the generation of distinct approaches to target RBPs, including HuR, in cancer and other human pathologies [210].
The downregulation of HuR function may be achieved through different strategies, with small molecules being the most popular class of inhibitors for this RBP. Small molecules, directly, inhibit HuR interaction with target RNAs, oligomerization, nucleocytoplasmic shuttling, or post-translational modification, further compromising its function. Dehydromutactin, MS-444, and okicenone are three low-molecular-weight molecules, found to inhibit both HuR dimerization and RNA binding, during a screening of compounds derived from Actinomyces cultures [172]. Among them, MS-444 showed the highest affinity and has proven to affect the viability of different cell lines [173,211,212,213]. Dihydrotanshinone-I (DHTS) [214], b40, and quercetin [215], as well as the coumarin derivative CMLD-2 [216,217,218], were, also, identified as chemical disruptors of the interaction between HuR and its target mRNAs. Other small-molecule inhibitors preventing HuR interaction with its target mRNAs include KH-3, which has been found to be promising for the treatment of breast cancer [219]. A new inhibitor is the muscone derivative ZM-32, a synthetic compound that shows antiangiogenic effects in breast cancer tumor cells, by blocking HuR association with VEGF and MMP9 mRNAs [220].
Except for ZM-32, which was tested in the human hepatoma HepG2 cell line without obtaining very successful outcomes [220], surprisingly, none of the abovementioned molecules have been tested in hepatobiliary tumors. Currently, studies reporting the use of small-molecule HuR inhibitors to tackle HCC are limited (Table 1). For instance, resveratrol (RSV) increased HuR mRNA and protein-expression levels in human liver cancer cell lines, which helped to raise MAT2B and SIRT1 expression, by stabilizing their mRNAs at the 3′-UTR. Importantly, the induced HuR, SIRT1, and MATβ proteins interact and stabilize each other, while compromising the binding of MATβ to MATα2. Eventually, this leads to an increase in SAMe levels, which might favor cell growth suppression and apoptosis during liver cancer, upon treatment with RSV [221]. In another study, N-Benzylcantharidinamide impaired HuR translocation to the cytosol and decreased the stability of MMP-9 mRNA and its expression, further inhibiting the invasive potential in metastatic Hep3B cells [222]. Additionally, since the nucleocytoplasmic transport of HuR depends on the interaction of this RBP with cytoskeletal proteins, the naturally occurring cytoskeletal inhibitors latrunculin A and blebbistatin have shown to exert antitumorigenic properties in human hepatoma cells, by interfering with the intracellular trafficking of HuR and its mRNA cargo [223]. Finally, the NEDDylation inhibitor Pevonedistat was shown to exert antitumoral effects in vitro and in vivo in liver cancer, partially through HuR destabilization. Importantly, overexpression of HuR in hepatoma cells offered resistance to pharmacological NEDDylation inhibition, while low levels of HuR sensitized cells to the treatment, suggesting that HuR levels determine the druggability of the NEDDylation pathway in HCC [224].
Notably, antisense oligonucleotides (ASOs) [225] and small interfering RNAs (siRNAs) exist for HuR. The latter have been widely used in the numerous studies mentioned throughout this review, in connection with HCC [187,188,189,194,197,199,204,206,208]. Related with siRNA technology as well, targeted delivery is, often, highly desirable and can be achieved thanks to the use of nanotechnology, for example, by means of the folate-receptor-targeted nanoparticle delivery of HuR siRNA, to reach lung cancer cells [226].

4. Future Perspectives

Herein, we have reviewed the current knowledge on the role of HuR in hepatobiliary tumors, including HCC and CCA. Considering increased HuR protein expression, cytoplasmic localization, and its relationship with patient outcome, HuR is regarded as a valuable diagnostic as well as a prognostic marker in HCC. Even though we recognize the growing attention of liquid biopsy in HCC, only invasive approaches have been carried out, to address the diagnosis and prognosis of HuR in these types of tumors, to date. With regard to CCA, high cytoplasmic expression and elevated tumor-gene levels of HuR have been described. However, special care should be taken when using cytological and pathological approaches, due to the highly desmoplastic nature of these types of tumors. In fact, in biliary tract carcinomas, biopsy samples are often inadequate for molecular profiling. Additionally, tissue sampling has reported high specificity but low sensitivity in diagnosis. Thereby, to further validate the relevance of HuR as a diagnostic and prognostic marker in CCA, further in vitro studies and in vivo pre-clinical mouse models are required.
Even though the role of HuR in CCA remains, largely, unexplored, novel mechanisms underlying HuR regulation have been addressed in HCC, with special focus on the importance of the PTM NEDDylation in the stabilization of HuR, preventing it from proteasome-mediated degradation [187]. Consequently, the relevance of HuR in pathological conditions, including cancer, has gained interest, with the advent of many naturally occurring and synthetic inhibitors of HuR. Nevertheless, given the lack of studies, using the currently available HuR inhibitors for the treatment of hepatobiliary tumors, and considering the heterogenous nature of HuR as a target affecting multiple pathways in many types of human cancers, it is, somehow, expected that the inhibition of HuR may be associated with undesired side-effects. Indeed, it has been described that HuR whole-body knock-out leads to embryonic lethality in mice, suggesting that HuR is, most probably, involved in regulating the fate of mRNAs encoding proteins implicated in key processes, such as organ development and tissue homeostasis [227].
Therefore, to take advantage of HuR as a therapeutic target, we propose that it would be critical to, specifically, inhibit the diverse interactions of HuR with its transcripts, which are uniquely relevant in the context of hepatobiliary cancers. For this purpose, the first step would be to interrogate the HuR RNA-binding signature in hepatobiliary tumors, which could be achieved, for example, by using RNA sequencing (RNA-Seq) approaches after the immunoprecipitation (IP) of the RNAs interacting with HuR, in the tumor and adjacent tissues, of large cohorts of HCC and CCA patients. Additionally, considering the relevance of HuR PTMs in liver disease, the screening of HuR PTM signature, including the residues susceptible of modification in liver cancer can unravel new unexpected and druggable regulatory mechanisms of HuR function. The screening of novel HuR RNA targets and PTMs in liver cancer as well as their clustering, according to the degree of liver disease, may, eventually, reveal subtypes of patients where certain HuR-related therapeutic approaches may be more suitable, paving the way for a more-personalized medicine. In other words, from this population-based study, we could, potentially, treat liver cancer by inhibiting the interactions of HuR either with (1) specific target RNAs or (2) peptides responsible for its PTM, which are ascribed to liver cancer. Indeed, short RNAs that compete with HuR for the binding to its target mRNAs, without affecting other HuR-mRNA binding interactions, had been previously suggested as a novel strategy for the management of HCC [228]. In this context, aptamers have, recently, emerged as small single-stranded DNA or RNA molecules that fold in a specific way, which is optimal to interact with a given target [229]. Thus, after elucidating HuR RNA targets and its PTMs in the context of liver cancer, we believe it would be possible to obtain aptamers blocking HuR interaction with both RNA and the peptides associated with PTMs, in a highly specific manner, minimizing potential adverse effects. This might be achieved through the screening of aptamer libraries, using the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) selection method. For this purpose, the understanding of HuR protein structure is of great benefit to predict whether the conformation of the aptamer would be suitable to inhibit the binding of HuR to its target.

5. Concluding Remarks

In conclusion, even though, in the past few years, great efforts have been carried out, in order to better understand the function of HuR and its regulation in hepatobiliary tumors, further studies are necessary to address the therapeutic potential of HuR inhibitors or, alternatively, a more-personalized treatment based on the selected inhibition of HuR and its RNA or the protein targets in HCC and CCA.

Author Contributions

Conceptualization, S.L.-O., T.C.D., B.B.-J., A.V.-C., I.D.-M. and M.L.M.-C.; writing—review and editing, S.L.-O., T.C.D., B.B.-J., A.V.-C., I.D.-M. and M.L.M.-C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Programa Retos-Colaboración, grant number RTC2019-007125-1 (to M.L.M.-C.); the Instituto de Salud Carlos III, Proyectos de Investigación en Salud, grant number DTS20/00138 (to M.L.M.-C.); the Departamento de Industria del Gobierno Vasco (to M.L.M.-C.); the Ministerio de Ciencia, Innovación y Universidades (MICINN), integrated in the Plan Estatal de Investigación Científica y Técnica e Innovación, grant numbers PID2020-117116RB-I00 (to M.L.M.-C.) and PGC2018-096049-B-I00 (to I.D.-M.); the Gobierno de Andalucía, grant numbers BIO-198, US-1254317, US-1257019, P18-FR-3487, and P18-HO-4091 (to I.D.-M.); the Universidad de Sevilla (to I.D.-M.); Fundación Ramón Areces (to I.D.-M.); co-funded by the Fondo Europeo de Desarrollo Regional (FEDER) (to MLM-C and I.D.-M.); and the La Caixa Foundation Program, grant number HR17-00601 (to M.L.M.-C.). We acknowledge investigator contracts granted by the Ministerio de Educación, Cultura y Deporte, grant number FPU016/01513 (to A.V.C); the Asociación Española Contra el Cáncer (AECC) (to T.C.-D.); and Gobierno Vasco (to S.L.-O.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hinman, M.N.; Lou, H. Diverse Molecular Functions of Hu Proteins. Cell. Mol. Life Sci. 2008, 65, 3168–3181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Simone, L.E.; Keene, J.D. Mechanisms Coordinating ELAV/Hu MRNA Regulons. Curr. Opin. Genet. Dev. 2013, 23, 35–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-Based Map of the Human Proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
  4. Scheiba, R.M.; Aroca, A.; Díaz-Moreno, I. HuR Thermal Stability Is Dependent on Domain Binding and upon Phosphorylation. Eur. Biophys. J. 2012, 41, 597–605. [Google Scholar] [CrossRef] [Green Version]
  5. Wang, H.; Zeng, F.; Liu, Q.; Liu, H.; Liu, Z.; Niu, L.; Teng, M.; Li, X. The Structure of the ARE-Binding Domains of Hu Antigen R (HuR) Undergoes Conformational Changes during RNA Binding. Acta Crystallogr. D Biol. Crystallogr. 2013, 69, 373–380. [Google Scholar] [CrossRef] [PubMed]
  6. Mujo, A.; Lixa, C.; Carneiro, L.A.M.; Anobom, C.D.; Almeida, F.C.; Pinheiro, A.S. (1)H, (15)N and (13)C Resonance Assignments of the RRM1 Domain of the Key Post-Transcriptional Regulator HuR. Biomol. NMR Assign. 2015, 9, 281–284. [Google Scholar] [CrossRef]
  7. Benoit, R.M.; Meisner, N.-C.; Kallen, J.; Graff, P.; Hemmig, R.; Cèbe, R.; Ostermeier, C.; Widmer, H.; Auer, M. The X-Ray Crystal Structure of the First RNA Recognition Motif and Site-Directed Mutagenesis Suggest a Possible HuR Redox Sensing Mechanism. J. Mol. Biol. 2010, 397, 1231–1244. [Google Scholar] [CrossRef]
  8. Poganik, J.R.; Van Hall-Beauvais, A.K.; Long, M.J.C.; Disare, M.T.; Zhao, Y.; Aye, Y. The MRNA-Binding Protein HuR Is a Kinetically-Privileged Electrophile Sensor. Helv. Chim. Acta 2020, 103, e2000041. [Google Scholar] [CrossRef]
  9. Fan, X.C.; Steitz, J.A. Overexpression of HuR, a Nuclear–Cytoplasmic Shuttling Protein, Increases the in Vivo Stability of ARE-Containing MRNAs. EMBO J. 1998, 17, 3448–3460. [Google Scholar] [CrossRef] [Green Version]
  10. Grammatikakis, I.; Abdelmohsen, K.; Gorospe, M. Posttranslational Control of HuR Function. Wiley Interdiscip. Rev. RNA 2017, 8, e1372. [Google Scholar] [CrossRef] [Green Version]
  11. Velázquez-Cruz, A.; Baños-Jaime, B.; Díaz-Quintana, A.; De la Rosa, M.A.; Díaz-Moreno, I. Post-Translational Control of RNA-Binding Proteins and Disease-Related Dysregulation. Front. Mol. Biosci. 2021, 8, 658852. [Google Scholar] [CrossRef] [PubMed]
  12. Scheiba, R.M.; de Opakua, A.I.; Díaz-Quintana, A.; Cruz-Gallardo, I.; Martínez-Cruz, L.A.; Martínez-Chantar, M.L.; Blanco, F.J.; Díaz-Moreno, I. The C-Terminal RNA Binding Motif of HuR is a Multi-Functional Domain Leading to HuR Oligomerization and Binding to U-Rich RNA Targets. RNA Biol. 2014, 11, 1250–1261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Díaz-Quintana, A.; García-Mauriño, S.M.; Díaz-Moreno, I. Dimerization Model of the C-Terminal RNA Recognition Motif of HuR. FEBS Lett. 2015, 589, 1059–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lixa, C.; Mujo, A.; de Magalhães, M.T.Q.; Almeida, F.C.L.; Lima, L.M.T.R.; Pinheiro, A.S. Oligomeric Transition and Dynamics of RNA Binding by the HuR RRM1 Domain in Solution. J. Biomol. NMR 2018, 72, 179–192. [Google Scholar] [CrossRef] [PubMed]
  15. Pabis, M.; Popowicz, G.M.; Stehle, R.; Fernández-Ramos, D.; Asami, S.; Warner, L.; García-Mauriño, S.M.; Schlundt, A.; Martínez-Chantar, M.L.; Díaz-Moreno, I.; et al. HuR Biological Function Involves RRM3-Mediated Dimerization and RNA Binding by All Three RRMs. Nucleic Acids Res. 2019, 47, 1011–1029. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, X.; Tanaka Hall, T.M. Structural Basis for Recognition of AU-Rich Element RNA by the HuD Protein. Nat. Struct. Biol. 2001, 8, 141–145. [Google Scholar] [CrossRef]
  17. Corley, M.; Burns, M.C.; Yeo, G.W. How RNA-Binding Proteins Interact with RNA: Molecules and Mechanisms. Mol. Cell 2020, 78, 9–29. [Google Scholar] [CrossRef]
  18. Della Volpe, S.; Nasti, R.; Queirolo, M.; Unver, M.Y.; Jumde, V.K.; Dömling, A.; Vasile, F.; Potenza, D.; Ambrosio, F.A.; Costa, G.; et al. Novel Compounds Targeting the RNA-Binding Protein HuR. Structure-Based Design, Synthesis, and Interaction Studies. ACS Med. Chem. Lett. 2019, 10, 615–620. [Google Scholar] [CrossRef]
  19. Mukherjee, N.; Corcoran, D.L.; Nusbaum, J.D.; Reid, D.W.; Georgiev, S.; Hafner, M.; Ascano, M.J.; Tuschl, T.; Ohler, U.; Keene, J.D. Integrative Regulatory Mapping Indicates That the RNA-Binding Protein HuR Couples Pre-MRNA Processing and MRNA Stability. Mol. Cell 2011, 43, 327–339. [Google Scholar] [CrossRef] [Green Version]
  20. Lebedeva, S.; Jens, M.; Theil, K.; Schwanhäusser, B.; Selbach, M.; Landthaler, M.; Rajewsky, N. Transcriptome-Wide Analysis of Regulatory Interactions of the RNA-Binding Protein HuR. Mol. Cell 2011, 43, 340–352. [Google Scholar] [CrossRef] [Green Version]
  21. Izquierdo, J.M. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Gallouzi, I.E.; Steitz, J.A. Delineation of MRNA Export Pathways by the Use of Cell-Permeable Peptides. Science 2001, 294, 1895–1901. [Google Scholar] [CrossRef] [PubMed]
  23. Prechtel, A.T.; Chemnitz, J.; Schirmer, S.; Ehlers, C.; Langbein-Detsch, I.; Stülke, J.; Dabauvalle, M.-C.; Kehlenbach, R.H.; Hauber, J. Expression of CD83 Is Regulated by HuR via a Novel Cis-Active Coding Region RNA Element. J. Biol. Chem. 2006, 281, 10912–10925. [Google Scholar] [CrossRef] [Green Version]
  24. Doller, A.; Akool, E.-S.; Huwiler, A.; Müller, R.; Radeke, H.H.; Pfeilschifter, J.; Eberhardt, W. Posttranslational Modification of the AU-Rich Element Binding Protein HuR by Protein Kinase Cdelta Elicits Angiotensin II-Induced Stabilization and Nuclear Export of Cyclooxygenase 2 MRNA. Mol. Cell. Biol. 2008, 28, 2608–2625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yi, J.; Chang, N.; Liu, X.; Guo, G.; Xue, L.; Tong, T.; Gorospe, M.; Wang, W. Reduced Nuclear Export of HuR MRNA by HuR Is Linked to the Loss of HuR in Replicative Senescence. Nucleic Acids Res. 2010, 38, 1547–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Rebane, A.; Aab, A.; Steitz, J.A. Transportins 1 and 2 Are Redundant Nuclear Import Factors for HnRNP A1 and HuR. RNA 2004, 10, 590–599. [Google Scholar] [CrossRef] [Green Version]
  27. Gallouzi, I.E.; Brennan, C.M.; Steitz, J.A. Protein Ligands Mediate the CRM1-Dependent Export of HuR in Response to Heat Shock. RNA 2001, 7, 1348–1361. [Google Scholar] [CrossRef] [Green Version]
  28. Abdelmohsen, K.; Gorospe, M. Posttranscriptional Regulation of Cancer Traits by HuR. Wiley Interdiscip. Rev. RNA 2010, 1, 214–229. [Google Scholar] [CrossRef]
  29. Srikantan, S.; Gorospe, M. HuR Function in Disease. Front. Biosci. Landmark Ed. 2012, 17, 189–205. [Google Scholar] [CrossRef] [Green Version]
  30. Kakuguchi, W.; Kitamura, T.; Kuroshima, T.; Ishikawa, M.; Kitagawa, Y.; Totsuka, Y.; Shindoh, M.; Higashino, F. HuR Knockdown Changes the Oncogenic Potential of Oral Cancer Cells. Mol. Cancer Res. 2010, 8, 520–528. [Google Scholar] [CrossRef] [Green Version]
  31. Giles, K.M.; Daly, J.M.; Beveridge, D.J.; Thomson, A.M.; Voon, D.C.; Furneaux, H.M.; Jazayeri, J.A.; Leedman, P.J. The 3’-Untranslated Region of P21WAF1 MRNA Is a Composite Cis-Acting Sequence Bound by RNA-Binding Proteins from Breast Cancer Cells, Including HuR and Poly(C)-Binding Protein. J. Biol. Chem. 2003, 278, 2937–2946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Wang, W.; Furneaux, H.; Cheng, H.; Caldwell, M.C.; Hutter, D.; Liu, Y.; Holbrook, N.; Gorospe, M. HuR Regulates P21 MRNA Stabilization by UV Light. Mol. Cell. Biol. 2000, 20, 760–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lafarga, V.; Cuadrado, A.; Lopez de Silanes, I.; Bengoechea, R.; Fernandez-Capetillo, O.; Nebreda, A.R. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wang, W.; Caldwell, M.C.; Lin, S.; Furneaux, H.; Gorospe, M. HuR Regulates Cyclin A and Cyclin B1 MRNA Stability during Cell Proliferation. EMBO J. 2000, 19, 2340–2350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Guo, X.; Hartley, R.S. HuR Contributes to Cyclin E1 Deregulation in MCF-7 Breast Cancer Cells. Cancer Res. 2006, 66, 7948–7956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Guo, X.; Wu, Y.; Hartley, R.S. Cold-Inducible RNA-Binding Protein Contributes to Human Antigen R and Cyclin E1 Deregulation in Breast Cancer. Mol. Carcinog. 2010, 49, 130–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Durie, D.; Lewis, S.M.; Liwak, U.; Kisilewicz, M.; Gorospe, M.; Holcik, M. RNA-Binding Protein HuR Mediates Cytoprotection through Stimulation of XIAP Translation. Oncogene 2011, 30, 1460–1469. [Google Scholar] [CrossRef] [Green Version]
  38. Cok, S.J.; Acton, S.J.; Morrison, A.R. The Proximal Region of the 3’-Untranslated Region of Cyclooxygenase-2 Is Recognized by a Multimeric Protein Complex Containing HuR, TIA-1, TIAR, and the Heterogeneous Nuclear Ribonucleoprotein U. J. Biol. Chem. 2003, 278, 36157–36162. [Google Scholar] [CrossRef] [Green Version]
  39. Linker, K.; Pautz, A.; Fechir, M.; Hubrich, T.; Greeve, J.; Kleinert, H. Involvement of KSRP in the Post-Transcriptional Regulation of Human INOS Expression-Complex Interplay of KSRP with TTP and HuR. Nucleic Acids Res. 2005, 33, 4813–4827. [Google Scholar] [CrossRef] [Green Version]
  40. Tschernatsch, M.M.O.; Mlecnik, B.; Trajanoski, Z.; Zechner, R.; Zimmermann, R. LPL-Mediated Lipolysis of VLDL Induces an Upregulation of AU-Rich MRNAs and an Activation of HuR in Endothelial Cells. Atherosclerosis 2006, 189, 310–317. [Google Scholar] [CrossRef]
  41. Esnault, S.; Malter, J.S. Hyaluronic Acid or TNF-Alpha plus Fibronectin Triggers Granulocyte Macrophage-Colony-Stimulating Factor MRNA Stabilization in Eosinophils yet Engages Differential Intracellular Pathways and MRNA Binding Proteins. J. Immunol. 2003, 171, 6780–6787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Topisirovic, I.; Siddiqui, N.; Orolicki, S.; Skrabanek, L.A.; Tremblay, M.; Hoang, T.; Borden, K.L.B. Stability of Eukaryotic Translation Initiation Factor 4E MRNA Is Regulated by HuR, and This Activity Is Dysregulated in Cancer. Mol. Cell. Biol. 2009, 29, 1152–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ghosh, M.; Aguila, H.L.; Michaud, J.; Ai, Y.; Wu, M.-T.; 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. Investig. 2009, 119, 3530–3543. [Google Scholar] [CrossRef] [PubMed]
  44. Masuda, K.; Abdelmohsen, K.; Kim, M.M.; Srikantan, S.; Lee, E.K.; Tominaga, K.; Selimyan, R.; Martindale, J.L.; Yang, X.; Lehrmann, E.; et al. Global Dissociation of HuR-MRNA Complexes Promotes Cell Survival after Ionizing Radiation. EMBO J. 2011, 30, 1040–1053. [Google Scholar] [CrossRef] [PubMed]
  45. Nabors, L.B.; Gillespie, G.Y.; Harkins, L.; King, P.H. 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. [Google Scholar] [PubMed]
  46. Amadio, M.; Bucolo, C.; Leggio, G.M.; Drago, F.; Govoni, S.; Pascale, A. The PKCbeta/HuR/VEGF Pathway in Diabetic Retinopathy. Biochem. Pharmacol. 2010, 80, 1230–1237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Avivi, A.; Shams, I.; Joel, A.; Lache, O.; Levy, A.P.; Nevo, E. Increased Blood Vessel Density Provides the Mole Rat Physiological Tolerance to Its Hypoxic Subterranean Habitat. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2005, 19, 1314–1316. [Google Scholar] [CrossRef]
  48. Li, X.; Lu, L.; Bush, D.J.; Zhang, X.; Zheng, L.; Suswam, E.A.; King, P.H. Mutant Copper-Zinc Superoxide Dismutase Associated with Amyotrophic Lateral Sclerosis Binds to Adenine/Uridine-Rich Stability Elements in the Vascular Endothelial Growth Factor 3’-Untranslated Region. J. Neurochem. 2009, 108, 1032–1044. [Google Scholar] [CrossRef] [Green Version]
  49. Lu, L.; Zheng, L.; Viera, L.; Suswam, E.; Li, Y.; Li, X.; Estévez, A.G.; King, P.H. Mutant Cu/Zn-Superoxide Dismutase Associated with Amyotrophic Lateral Sclerosis Destabilizes Vascular Endothelial Growth Factor MRNA and Downregulates Its Expression. J. Neurosci. 2007, 27, 7929–7938. [Google Scholar] [CrossRef] [Green Version]
  50. Basu, A.; Datta, D.; Zurakowski, D.; Pal, S. Altered VEGF MRNA Stability Following Treatments with Immunosuppressive Agents: Implications for Cancer Development. J. Biol. Chem. 2010, 285, 25196–25202. [Google Scholar] [CrossRef] [Green Version]
  51. Abdelmohsen, K.; Pullmann, R.J.; Lal, A.; Kim, H.H.; Galban, S.; Yang, X.; Blethrow, J.D.; Walker, M.; Shubert, J.; Gillespie, D.A.; et al. Phosphorylation of HuR by Chk2 Regulates SIRT1 Expression. Mol. Cell 2007, 25, 543–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Niesporek, S.; Kristiansen, G.; Thoma, A.; Weichert, W.; Noske, A.; Buckendahl, A.-C.; 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. [Google Scholar] [CrossRef] [PubMed]
  53. Suzuki, E.; Tsutsumi, A.; Sugihara, M.; Mamura, M.; Goto, D.; Matsumoto, I.; Ito, S.; Ikeda, K.; Ochiai, N.; Sato, Y.; et al. Expression of TNF-Alpha, Tristetraprolin, T-Cell Intracellular Antigen-1 and Hu Antigen R Genes in Synovium of Patients with Rheumatoid Arthritis. Int. J. Mol. Med. 2006, 18, 273–278. [Google Scholar] [PubMed]
  54. Abdelmohsen, K.; Lal, A.; Kim, H.H.; Gorospe, M. Posttranscriptional Orchestration of an Anti-Apoptotic Program by HuR. Cell Cycle 2007, 6, 1288–1292. [Google Scholar] [CrossRef] [PubMed]
  55. Ishimaru, D.; Ramalingam, S.; Sengupta, T.K.; Bandyopadhyay, S.; Dellis, S.; Tholanikunnel, B.G.; Fernandes, D.J.; Spicer, E.K. Regulation of Bcl-2 Expression by HuR in HL60 Leukemia Cells and A431 Carcinoma Cells. Mol. Cancer Res. 2009, 7, 1354–1366. [Google Scholar] [CrossRef] [Green Version]
  56. Bandyopadhyay, S.; Sengupta, T.K.; Spicer, E.K. PMA Induces Stabilization of Oncostatin M MRNA in Human Lymphoma U937 Cells. Biochem. J. 2008, 410, 177–186. [Google Scholar] [CrossRef]
  57. Dixon, D.A.; Tolley, N.D.; King, P.H.; Nabors, L.B.; McIntyre, T.M.; Zimmerman, G.A.; Prescott, S.M. Altered Expression of the MRNA Stability Factor HuR Promotes Cyclooxygenase-2 Expression in Colon Cancer Cells. J. Clin. Investig. 2001, 108, 1657–1665. [Google Scholar] [CrossRef]
  58. Lu, Z.; Luo, R.Z.; Peng, H.; Rosen, D.G.; Atkinson, E.N.; Warneke, C.; Huang, M.; Nishmoto, A.; Liu, J.; Liao, W.S.-L.; et al. Transcriptional and Posttranscriptional Down-Regulation of the Imprinted Tumor Suppressor Gene ARHI (DRAS3) in Ovarian Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 2404–2413. [Google Scholar] [CrossRef] [Green Version]
  59. Young, L.E.; Sanduja, S.; Bemis-Standoli, K.; Pena, E.A.; Price, R.L.; Dixon, D.A. The MRNA Binding Proteins HuR and Tristetraprolin Regulate Cyclooxygenase 2 Expression during Colon Carcinogenesis. Gastroenterology 2009, 136, 1669–1679. [Google Scholar] [CrossRef] [Green Version]
  60. Erkinheimo, T.-L.; Lassus, H.; Sivula, A.; Sengupta, S.; Furneaux, H.; Hla, T.; Haglund, C.; Butzow, R.; Ristimäki, A. Cytoplasmic HuR Expression Correlates with Poor Outcome and with Cyclooxygenase 2 Expression in Serous Ovarian Carcinoma. Cancer Res. 2003, 63, 7591–7594. [Google Scholar]
  61. Barbisan, F.; Mazzucchelli, R.; Santinelli, A.; Lopez-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. 2009, 56, 105–112. [Google Scholar] [CrossRef]
  62. Mrena, J.; Wiksten, J.-P.; 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. Off. J. Am. Assoc. Cancer Res. 2005, 11, 7362–7368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Song, I.-S.; Tatebe, S.; Dai, W.; Kuo, M.T. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Farooq, F.; Balabanian, S.; Liu, X.; Holcik, M.; MacKenzie, A. P38 Mitogen-Activated Protein Kinase Stabilizes SMN MRNA through RNA Binding Protein HuR. Hum. Mol. Genet. 2009, 18, 4035–4045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Okamoto, S.; Ji, H.; Howie, D.; Clarke, K.; Gullo, C.; Manning, S.; Coyle, A.J.; Terhorst, C. Expression of the SH2D1A Gene Is Regulated by a Combination of Transcriptional and Post-Transcriptional Mechanisms. Eur. J. Immunol. 2004, 34, 3176–3186. [Google Scholar] [CrossRef]
  66. Li, F.; Hu, D.Y.; Liu, S.; Mahavadi, S.; Yen, W.; Murthy, K.S.; Khalili, K.; Hu, W. RNA-Binding Protein HuR Regulates RGS4 MRNA Stability in Rabbit Colonic Smooth Muscle Cells. Am. J. Physiol. Cell Physiol. 2010, 299, C1418–C1429. [Google Scholar] [CrossRef] [Green Version]
  67. Danilin, S.; Sourbier, C.; Thomas, L.; Rothhut, S.; Lindner, V.; Helwig, J.-J.; Jacqmin, D.; Lang, H.; Massfelder, T. Von Hippel-Lindau Tumor Suppressor Gene-Dependent MRNA Stabilization of the Survival Factor Parathyroid Hormone-Related Protein in Human Renal Cell Carcinoma by the RNA-Binding Protein HuR. Carcinogenesis 2009, 30, 387–396. [Google Scholar] [CrossRef] [Green Version]
  68. Drury, G.L.; Di Marco, S.; Dormoy-Raclet, V.; Desbarats, J.; Gallouzi, I.-E. FasL Expression in Activated T Lymphocytes Involves HuR-Mediated Stabilization. J. Biol. Chem. 2010, 285, 31130–31138. [Google Scholar] [CrossRef] [Green Version]
  69. Figueroa, A.; Cuadrado, A.; Fan, J.; Atasoy, U.; Muscat, G.E.; Muñoz-Canoves, P.; Gorospe, M.; Muñoz, A. Role of HuR in Skeletal Myogenesis through Coordinate Regulation of Muscle Differentiation Genes. Mol. Cell. Biol. 2003, 23, 4991–5004. [Google Scholar] [CrossRef] [Green Version]
  70. van der Giessen, K.; Di-Marco, S.; Clair, E.; Gallouzi, I.E. RNAi-Mediated HuR Depletion Leads to the Inhibition of Muscle Cell Differentiation. J. Biol. Chem. 2003, 278, 47119–47128. [Google Scholar] [CrossRef] [Green Version]
  71. Deschênes-Furry, J.; Bélanger, G.; Mwanjewe, J.; Lunde, J.A.; Parks, R.J.; Perrone-Bizzozero, N.; Jasmin, B.J. The RNA-Binding Protein HuR Binds to Acetylcholinesterase Transcripts and Regulates Their Expression in Differentiating Skeletal Muscle Cells. J. Biol. Chem. 2005, 280, 25361–25368. [Google Scholar] [CrossRef] [Green Version]
  72. Bronicki, L.M.; Jasmin, B.J. Trans-Acting Factors Governing Acetylcholinesterase MRNA Metabolism in Neurons. Front. Mol. Neurosci. 2012, 5, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Mazan-Mamczarz, K.; Galbán, S.; López de Silanes, I.; Martindale, J.L.; Atasoy, U.; Keene, J.D.; Gorospe, M. RNA-Binding Protein HuR Enhances P53 Translation in Response to Ultraviolet Light Irradiation. Proc. Natl. Acad. Sci. USA 2003, 100, 8354–8359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zou, T.; Mazan-Mamczarz, K.; Rao, J.N.; Liu, L.; Marasa, B.S.; Zhang, A.-H.; Xiao, L.; Pullmann, R.; Gorospe, M.; Wang, J.-Y. Polyamine Depletion Increases Cytoplasmic Levels of RNA-Binding Protein HuR Leading to Stabilization of Nucleophosmin and P53 MRNAs. J. Biol. Chem. 2006, 281, 19387–19394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. de Frutos, S.; Nitta, C.H.; Caldwell, E.; Friedman, J.; González Bosc, L.V. Regulation of Soluble Guanylyl Cyclase-Alpha1 Expression in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of NFATc3 and HuR. Am. J. Physiol. Lung Cell. Mol. Physiol. 2009, 297, L475–L486. [Google Scholar] [CrossRef] [Green Version]
  76. 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. [Google Scholar] [CrossRef] [Green Version]
  77. Haeussler, J.; Haeusler, J.; Striebel, A.M.; Assum, G.; Vogel, W.; Furneaux, H.; Krone, W. Tumor Antigen HuR Binds Specifically to One of Five Protein-Binding Segments in the 3’-Untranslated Region of the Neurofibromin Messenger RNA. Biochem. Biophys. Res. Commun. 2000, 267, 726–732. [Google Scholar] [CrossRef]
  78. Abdelmohsen, K.; Srikantan, S.; Yang, X.; Lal, A.; Kim, H.H.; Kuwano, Y.; Galban, S.; Becker, K.G.; Kamara, D.; de Cabo, R.; et al. Ubiquitin-Mediated Proteolysis of HuR by Heat Shock. EMBO J. 2009, 28, 1271–1282. [Google Scholar] [CrossRef] [Green Version]
  79. Lin, F.-Y.; Chen, Y.-H.; Lin, Y.-W.; Tsai, J.-S.; Chen, J.-W.; Wang, H.-J.; Chen, Y.-L.; Li, C.-Y.; Lin, S.-J. The Role of Human Antigen R, an RNA-Binding Protein, in Mediating the Stabilization of Toll-like Receptor 4 MRNA Induced by Endotoxin: A Novel Mechanism Involved in Vascular Inflammation. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 2622–2629. [Google Scholar] [CrossRef] [Green Version]
  80. Dong, R.; Lu, J.-G.; Wang, Q.; He, X.-L.; Chu, Y.-K.; Ma, Q.-J. Stabilization of Snail by HuR in the Process of Hydrogen Peroxide Induced Cell Migration. Biochem. Biophys. Res. Commun. 2007, 356, 318–321. [Google Scholar] [CrossRef]
  81. Annabi, B.; Bouzeghrane, M.; Currie, J.-C.; Dulude, H.; Daigneault, L.; Garde, S.; Rabbani, S.A.; Panchal, C.; Wu, J.J.; Béliveau, R. Inhibition of MMP-9 Secretion by the Anti-Metastatic PSP94-Derived Peptide PCK3145 Requires Cell Surface Laminin Receptor Signaling. Anticancer. Drugs 2006, 17, 429–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Annabi, B.; Currie, J.-C.; Moghrabi, A.; Béliveau, R. Inhibition of HuR and MMP-9 Expression in Macrophage-Differentiated HL-60 Myeloid Leukemia Cells by Green Tea Polyphenol EGCg. Leuk. Res. 2007, 31, 1277–1284. [Google Scholar] [CrossRef] [PubMed]
  83. Krishnamurthy, P.; Rajasingh, J.; Lambers, E.; Qin, G.; Losordo, D.W.; Kishore, R. IL-10 Inhibits Inflammation and Attenuates Left Ventricular Remodeling after Myocardial Infarction via Activation of STAT3 and Suppression of HuR. Circ. Res. 2009, 104, e9–e18. [Google Scholar] [CrossRef] [PubMed]
  84. Akool, E.-S.; Kleinert, H.; Hamada, F.M.A.; Abdelwahab, M.H.; Förstermann, 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. [Google Scholar] [CrossRef] [Green Version]
  85. Huwiler, A.; Akool, e.-S.; Aschrafi, A.; Hamada, F.M.A.; 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, 51758–51769. [Google Scholar] [CrossRef] [Green Version]
  86. Woo, H.-H.; Zhou, Y.; Yi, X.; David, C.L.; Zheng, W.; Gilmore-Hebert, M.; Kluger, H.M.; Ulukus, E.C.; Baker, T.; Stoffer, J.B.; et al. Regulation of Non-AU-Rich Element Containing c-Fms Proto-Oncogene Expression by HuR in Breast Cancer. Oncogene 2009, 28, 1176–1186. [Google Scholar] [CrossRef] [Green Version]
  87. Kuwano, Y.; Kim, H.H.; Abdelmohsen, K.; Pullmann, R.J.; Martindale, J.L.; 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. [Google Scholar] [CrossRef] [Green Version]
  88. Wang, J.G.; Collinge, M.; Ramgolam, V.; Ayalon, O.; Fan, X.C.; Pardi, R.; Bender, J.R. LFA-1-Dependent HuR Nuclear Export and Cytokine MRNA Stabilization in T Cell Activation. J. Immunol. 2006, 176, 2105–2113. [Google Scholar] [CrossRef] [Green Version]
  89. Ming, X.F.; 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. [Google Scholar] [CrossRef] [Green Version]
  90. Yarovinsky, T.O.; Butler, N.S.; Monick, M.M.; Hunninghake, G.W. Early Exposure to IL-4 Stabilizes IL-4 MRNA in CD4+ T Cells via RNA-Binding Protein HuR. J. Immunol. 2006, 177, 4426–4435. [Google Scholar] [CrossRef] [Green Version]
  91. Gealy, C.; Denson, M.; Humphreys, C.; McSharry, B.; Wilkinson, G.; Caswell, R. Posttranscriptional Suppression of Interleukin-6 Production by Human Cytomegalovirus. J. Virol. 2005, 79, 472–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zhou, H.; Jarujaron, S.; Gurley, E.C.; Chen, L.; Ding, H.; Studer, E.; Pandak, W.M.J.; Hu, W.; Zou, T.; Wang, J.-Y.; et al. HIV Protease Inhibitors Increase TNF-Alpha and IL-6 Expression in Macrophages: Involvement of the RNA-Binding Protein HuR. Atherosclerosis 2007, 195, e134–e143. [Google Scholar] [CrossRef] [PubMed]
  93. Suswam, E.A.; Nabors, L.B.; Huang, Y.; Yang, X.; King, P.H. 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. [Google Scholar] [CrossRef] [PubMed]
  94. Pullmann Jr, R.; Kim, H.H.; Abdelmohsen, K.; Lal, A.; Martindale, J.L.; Yang, X.; Gorospe, M. Analysis of Turnover and Translation Regulatory RNA-Binding Protein Expression through Binding to Cognate MRNAs. Mol. Cell. Biol. 2007, 27, 6265–6278. [Google Scholar] [CrossRef] [Green Version]
  95. Al-Ahmadi, W.; Al-Ghamdi, M.; Al-Haj, L.; Al-Saif, M.; Khabar, K.S.A. Alternative Polyadenylation Variants of the RNA Binding Protein, HuR: Abundance, Role of AU-Rich Elements and Auto-Regulation. Nucleic Acids Res. 2009, 37, 3612–3624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Mrena, J.; Wiksten, J.-P.; Kokkola, A.; Nordling, S.; Haglund, C.; Ristimäki, A. Prognostic Significance of Cyclin A in Gastric Cancer. Int. J. Cancer 2006, 119, 1897–1901. [Google Scholar] [CrossRef]
  97. 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. [Google Scholar] [CrossRef]
  98. Galbán, S.; Kuwano, Y.; Pullmann, R.J.; Martindale, J.L.; Kim, H.H.; Lal, A.; Abdelmohsen, K.; Yang, X.; Dang, Y.; Liu, J.O.; et al. RNA-Binding Proteins HuR and PTB Promote the Translation of Hypoxia-Inducible Factor 1alpha. Mol. Cell. Biol. 2008, 28, 93–107. [Google Scholar] [CrossRef] [Green Version]
  99. Ayupova, D.A.; Singh, M.; Leonard, E.C.; Basile, D.P.; Lee, B.S. Expression of the RNA-Stabilizing Protein HuR in Ischemia-Reperfusion Injury of Rat Kidney. Am. J. Physiol. Renal Physiol. 2009, 297, F95–F105. [Google Scholar] [CrossRef] [Green Version]
  100. 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. [Google Scholar] [CrossRef]
  101. Ido, K.; Nakagawa, T.; Sakuma, T.; Takeuchi, H.; Sato, K.; Kubota, T. Expression of Vascular Endothelial Growth Factor-A and MRNA Stability Factor HuR in Human Astrocytic Tumors. Neuropathology 2008, 28, 604–611. [Google Scholar] [CrossRef] [PubMed]
  102. Sakuma, T.; Nakagawa, T.; Ido, K.; Takeuchi, H.; Sato, K.; Kubota, T. Expression of Vascular Endothelial Growth Factor-A and MRNA Stability Factor HuR in Human Meningiomas. J. Neurooncol. 2008, 88, 143–155. [Google Scholar] [CrossRef] [PubMed]
  103. Mazan-Mamczarz, K.; Hagner, P.R.; Corl, S.; Srikantan, S.; Wood, W.H.; Becker, K.G.; Gorospe, M.; Keene, J.D.; Levenson, A.S.; Gartenhaus, R.B. Post-Transcriptional Gene Regulation by HuR Promotes a More Tumorigenic Phenotype. Oncogene 2008, 27, 6151–6163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Bhattacharyya, S.N.; Habermacher, R.; Martine, U.; Closs, E.I.; Filipowicz, W. Relief of MicroRNA-Mediated Translational Repression in Human Cells Subjected to Stress. Cell 2006, 125, 1111–1124. [Google Scholar] [CrossRef] [Green Version]
  105. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Kullmann, M.; Göpfert, 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. [Google Scholar] [CrossRef] [Green Version]
  107. Meng, Z.; King, P.H.; Nabors, L.B.; Jackson, N.L.; Chen, C.-Y.; Emanuel, P.D.; Blume, S.W. The ELAV RNA-Stability Factor HuR Binds the 5’-Untranslated Region of the Human IGF-IR Transcript and Differentially Represses Cap-Dependent and IRES-Mediated Translation. Nucleic Acids Res. 2005, 33, 2962–2979. [Google Scholar] [CrossRef] [Green Version]
  108. Yeh, C.-H.; Hung, L.-Y.; Hsu, C.; Le, S.-Y.; Lee, P.-T.; Liao, W.-L.; Lin, Y.-T.; Chang, W.-C.; Tseng, J.T. RNA-Binding Protein HuR Interacts with Thrombomodulin 5’untranslated Region and Represses Internal Ribosome Entry Site-Mediated Translation under IL-1 Beta Treatment. Mol. Biol. Cell 2008, 19, 3812–3822. [Google Scholar] [CrossRef] [Green Version]
  109. 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. [Google Scholar] [CrossRef] [Green Version]
  110. Kim, H.H.; Kuwano, Y.; Srikantan, S.; Lee, E.K.; Martindale, J.L.; Gorospe, M. HuR Recruits Let-7/RISC to Repress c-Myc Expression. Genes Dev. 2009, 23, 1743–1748. [Google Scholar] [CrossRef] [Green Version]
  111. Yoon, J.-H.; Abdelmohsen, K.; Kim, J.; Yang, X.; Martindale, J.L.; Tominaga-Yamanaka, K.; White, E.J.; Orjalo, A.V.; Rinn, J.L.; Kreft, S.G.; et al. Scaffold Function of Long Non-Coding RNA HOTAIR in Protein Ubiquitination. Nat. Commun. 2013, 4, 2939. [Google Scholar] [CrossRef] [PubMed]
  112. Lu, Y.-C.; Chang, S.-H.; Hafner, M.; Li, X.; Tuschl, T.; Elemento, O.; Hla, T. ELAVL1 Modulates Transcriptome-Wide MiRNA Binding in Murine Macrophages. Cell Rep. 2014, 9, 2330–2343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Poria, D.K.; Guha, A.; Nandi, I.; Ray, P.S. RNA-Binding Protein HuR Sequesters MicroRNA-21 to Prevent Translation Repression of Proinflammatory Tumor Suppressor Gene Programmed Cell Death 4. Oncogene 2016, 35, 1703–1715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Mukherjee, K.; Ghoshal, B.; Ghosh, S.; Chakrabarty, Y.; Shwetha, S.; Das, S.; Bhattacharyya, S.N. Reversible HuR-MicroRNA Binding Controls Extracellular Export of MiR-122 and Augments Stress Response. EMBO Rep. 2016, 17, 1184–1203. [Google Scholar] [CrossRef] [PubMed]
  115. Yoon, J.-H.; Abdelmohsen, K.; Srikantan, S.; Yang, X.; Martindale, J.L.; De, S.; Huarte, M.; Zhan, M.; Becker, K.G.; Gorospe, M. LincRNA-P21 Suppresses Target MRNA Translation. Mol. Cell 2012, 47, 648–655. [Google Scholar] [CrossRef] [Green Version]
  116. Chai, Y.; Liu, J.; Zhang, Z.; Liu, L. HuR-Regulated LncRNA NEAT1 Stability in Tumorigenesis and Progression of Ovarian Cancer. Cancer Med. 2016, 5, 1588–1598. [Google Scholar] [CrossRef]
  117. Zou, T.; Jaladanki, S.K.; Liu, L.; Xiao, L.; Chung, H.K.; Wang, J.-Y.; Xu, Y.; Gorospe, M.; Wang, J.-Y. H19 Long Noncoding RNA Regulates Intestinal Epithelial Barrier Function via MicroRNA 675 by Interacting with RNA-Binding Protein HuR. Mol. Cell. Biol. 2016, 36, 1332–1341. [Google Scholar] [CrossRef] [Green Version]
  118. Jeyaraj, S.C.; Singh, M.; Ayupova, D.A.; Govindaraju, S.; Lee, B.S. Transcriptional Control of Human Antigen R by Bone Morphogenetic Protein. J. Biol. Chem. 2010, 285, 4432–4440. [Google Scholar] [CrossRef] [Green Version]
  119. Kang, M.-J.; Ryu, B.-K.; Lee, M.-G.; Han, J.; Lee, J.-H.; Ha, T.-K.; Byun, D.-S.; Chae, K.-S.; Lee, B.-H.; Chun, H.S.; et al. NF-KappaB Activates Transcription of the RNA-Binding Factor HuR, via PI3K-AKT Signaling, to Promote Gastric Tumorigenesis. Gastroenterology 2008, 135, 2030–2042.e3. [Google Scholar] [CrossRef]
  120. Dai, W.; Zhang, G.; Makeyev, E.V. RNA-Binding Protein HuR Autoregulates Its Expression by Promoting Alternative Polyadenylation Site Usage. Nucleic Acids Res. 2012, 40, 787–800. [Google Scholar] [CrossRef] [Green Version]
  121. Singh, M.; Martinez, A.R.; Govindaraju, S.; Lee, B.S. HuR Inhibits Apoptosis by Amplifying Akt Signaling through a Positive Feedback Loop. J. Cell. Physiol. 2013, 228, 182–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Chang, N.; Ge, J.; Xiu, L.; Zhao, Z.; Duan, X.; Tian, L.; Xie, J.; Yang, L.; Li, L. HuR Mediates Motility of Human Bone Marrow-Derived Mesenchymal Stem Cells Triggered by Sphingosine 1-Phosphate in Liver Fibrosis. J. Mol. Med. 2017, 95, 69–82. [Google Scholar] [CrossRef] [PubMed]
  123. Bai, D.; Gao, Q.; Li, C.; Ge, L.; Gao, Y.; Wang, H. A Conserved TGFβ1/HuR Feedback Circuit Regulates the Fibrogenic Response in Fibroblasts. Cell. Signal. 2012, 24, 1426–1432. [Google Scholar] [CrossRef] [PubMed]
  124. Guha, A.; Nag, S.; Ray, P.S. Negative Feedback Regulation by HuR Controls TRIM21 Expression and Function in Response to UV Radiation. Sci. Rep. 2020, 10, 11753. [Google Scholar] [CrossRef]
  125. Guo, X.; Wu, Y.; Hartley, R.S. MicroRNA-125a Represses Cell Growth by Targeting HuR in Breast Cancer. RNA Biol. 2009, 6, 575–583. [Google Scholar] [CrossRef] [Green Version]
  126. Abdelmohsen, K.; Kim, M.M.; Srikantan, S.; Mercken, E.M.; Brennan, S.E.; Wilson, G.M.; de Cabo, R.; Gorospe, M. MiR-519 Suppresses Tumor Growth by Reducing HuR Levels. Cell Cycle 2010, 9, 1354–1359. [Google Scholar] [CrossRef] [Green Version]
  127. Leucci, E.; Zriwil, A.; Gregersen, L.H.; Jensen, K.T.; Obad, S.; Bellan, C.; Leoncini, L.; Kauppinen, S.; Lund, A.H. Inhibition of MiR-9 de-Represses HuR and DICER1 and Impairs Hodgkin Lymphoma Tumour Outgrowth in Vivo. Oncogene 2012, 31, 5081–5089. [Google Scholar] [CrossRef] [Green Version]
  128. Guo, J.; Li, M.; Meng, X.; Sui, J.; Dou, L.; Tang, W.; Huang, X.; Man, Y.; Wang, S.; Li, J. MiR-291b-3p Induces Apoptosis in Liver Cell Line NCTC1469 by Reducing the Level of RNA-Binding Protein HuR. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2014, 33, 810–822. [Google Scholar] [CrossRef]
  129. Roff, A.N.; Craig, T.J.; August, A.; Stellato, C.; Ishmael, F.T. MicroRNA-570-3p Regulates HuR and Cytokine Expression in Airway Epithelial Cells. Am. J. Clin. Exp. Immunol. 2014, 3, 68–83. [Google Scholar]
  130. Guo, X.; Connick, M.C.; Vanderhoof, J.; Ishak, M.-A.; Hartley, R.S. MicroRNA-16 Modulates HuR Regulation of Cyclin E1 in Breast Cancer Cells. Int. J. Mol. Sci. 2015, 16, 7112–7132. [Google Scholar] [CrossRef]
  131. Liu, Y.; Chen, X.; Cheng, R.; Yang, F.; Yu, M.; Wang, C.; Cui, S.; Hong, Y.; Liang, H.; Liu, M.; et al. The Jun/MiR-22/HuR Regulatory Axis Contributes to Tumourigenesis in Colorectal Cancer. Mol. Cancer 2018, 17, 11. [Google Scholar] [CrossRef] [PubMed]
  132. Al-Haidari, A.; Algaber, A.; Madhi, R.; Syk, I.; Thorlacius, H. MiR-155-5p Controls Colon Cancer Cell Migration via Post-Transcriptional Regulation of Human Antigen R (HuR). Cancer Lett. 2018, 421, 145–151. [Google Scholar] [CrossRef] [PubMed]
  133. Li, H.; Park, S.; Kilburn, B.; Jelinek, M.A.; Henschen-Edman, A.; Aswad, D.W.; Stallcup, M.R.; Laird-Offringa, I.A. Lipopolysaccharide-Induced Methylation of HuR, an MRNA-Stabilizing Protein, by CARM1. Coactivator-Associated Arginine Methyltransferase. J. Biol. Chem. 2002, 277, 44623–44630. [Google Scholar] [CrossRef] [Green Version]
  134. Calvanese, V.; Lara, E.; Suárez-Alvarez, B.; Abu Dawud, R.; Vázquez-Chantada, M.; Martínez-Chantar, M.L.; Embade, N.; López-Nieva, P.; Horrillo, A.; Hmadcha, A.; et al. Sirtuin 1 Regulation of Developmental Genes during Differentiation of Stem Cells. Proc. Natl. Acad. Sci. USA 2010, 107, 13736–13741. [Google Scholar] [CrossRef] [Green Version]
  135. Pang, L.; Tian, H.; Chang, N.; Yi, J.; Xue, L.; Jiang, B.; Gorospe, M.; Zhang, X.; Wang, W. Loss of CARM1 Is Linked to Reduced HuR Function in Replicative Senescence. BMC Mol. Biol. 2013, 14, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Vigouroux, C.; Casse, J.-M.; Battaglia-Hsu, S.-F.; Brochin, L.; Luc, A.; Paris, C.; Lacomme, S.; Gueant, J.-L.; Vignaud, J.-M.; Gauchotte, G. Methyl(R217)HuR and MCM6 Are Inversely Correlated and Are Prognostic Markers in Non Small Cell Lung Carcinoma. Lung Cancer 2015, 89, 189–196. [Google Scholar] [CrossRef] [PubMed]
  137. Filippova, N.; Yang, X.; Nabors, L.B. Growth Factor Dependent Regulation of Centrosome Function and Genomic Instability by HuR. Biomolecules 2015, 5, 263–281. [Google Scholar] [CrossRef] [Green Version]
  138. Yoon, J.-H.; Abdelmohsen, K.; Srikantan, S.; Guo, R.; Yang, X.; Martindale, J.L.; Gorospe, M. Tyrosine Phosphorylation of HuR by JAK3 Triggers Dissociation and Degradation of HuR Target MRNAs. Nucleic Acids Res. 2014, 42, 1196–1208. [Google Scholar] [CrossRef] [Green Version]
  139. Yu, T.-X.; Wang, P.-Y.; Rao, J.N.; Zou, T.; Liu, L.; Xiao, L.; Gorospe, M.; Wang, J.-Y. Chk2-Dependent HuR Phosphorylation Regulates Occludin MRNA Translation and Epithelial Barrier Function. Nucleic Acids Res. 2011, 39, 8472–8487. [Google Scholar] [CrossRef] [Green Version]
  140. Akaike, Y.; Masuda, K.; Kuwano, Y.; Nishida, K.; Kajita, K.; Kurokawa, K.; Satake, Y.; Shoda, K.; Imoto, I.; Rokutan, K. HuR Regulates Alternative Splicing of the TRA2β Gene in Human Colon Cancer Cells under Oxidative Stress. Mol. Cell. Biol. 2014, 34, 2857–2873. [Google Scholar] [CrossRef] [Green Version]
  141. Liu, L.; Rao, J.N.; Zou, T.; Xiao, L.; Wang, P.-Y.; Turner, D.J.; Gorospe, M.; Wang, J.-Y. Polyamines Regulate C-Myc Translation through Chk2-Dependent HuR Phosphorylation. Mol. Biol. Cell 2009, 20, 4885–4898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Liao, W.-L.; Wang, W.-C.; Chang, W.-C.; Tseng, J.T. The RNA-Binding Protein HuR Stabilizes Cytosolic Phospholipase A2α MRNA under Interleukin-1β Treatment in Non-Small Cell Lung Cancer A549 Cells. J. Biol. Chem. 2011, 286, 35499–35508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Doller, A.; Huwiler, A.; Müller, R.; Radeke, H.H.; 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. [Google Scholar] [CrossRef] [PubMed]
  144. Kim, H.H.; Abdelmohsen, K.; Lal, A.; Pullmann, R.J.; Yang, X.; Galban, S.; Srikantan, S.; Martindale, J.L.; Blethrow, J.; Shokat, K.M.; et al. Nuclear HuR Accumulation through Phosphorylation by Cdk1. Genes Dev. 2008, 22, 1804–1815. [Google Scholar] [CrossRef] [Green Version]
  145. Filippova, N.; Yang, X.; King, P.; Nabors, L.B. Phosphoregulation of the RNA-Binding Protein Hu Antigen R (HuR) by Cdk5 Affects Centrosome Function. J. Biol. Chem. 2012, 287, 32277–32287. [Google Scholar] [CrossRef] [Green Version]
  146. Kim, H.H.; Yang, X.; Kuwano, Y.; Gorospe, M. Modification at HuR(S242) Alters HuR Localization and Proliferative Influence. Cell Cycle 2008, 7, 3371–3377. [Google Scholar] [CrossRef]
  147. Chu, P.-C.; Chuang, H.-C.; Kulp, S.K.; Chen, C.-S. The MRNA-Stabilizing Factor HuR Protein Is Targeted by β-TrCP Protein for Degradation in Response to Glycolysis Inhibition. J. Biol. Chem. 2012, 287, 43639–43650. [Google Scholar] [CrossRef] [Green Version]
  148. Doller, A.; Winkler, C.; Azrilian, I.; Schulz, S.; Hartmann, S.; Pfeilschifter, J.; Eberhardt, W. High-Constitutive HuR Phosphorylation at Ser 318 by PKCδ Propagates Tumor Relevant Functions in Colon Carcinoma Cells. Carcinogenesis 2011, 32, 676–685. [Google Scholar] [CrossRef] [Green Version]
  149. Latorre, E.; Castiglioni, I.; Gatto, P.; Carelli, S.; Quattrone, A.; Provenzani, A. Loss of Protein Kinase Cδ/HuR Interaction Is Necessary to Doxorubicin Resistance in Breast Cancer Cell Lines. J. Pharmacol. Exp. Ther. 2014, 349, 99–106. [Google Scholar] [CrossRef] [Green Version]
  150. Ubersax, J.A.; Woodbury, E.L.; Quang, P.N.; Paraz, M.; Blethrow, J.D.; Shah, K.; Shokat, K.M.; Morgan, D.O. Targets of the Cyclin-Dependent Kinase Cdk1. Nature 2003, 425, 859–864. [Google Scholar] [CrossRef]
  151. Degese, M.S.; Tanos, T.; Naipauer, J.; Gingerich, T.; Chiappe, D.; Echeverria, P.; LaMarre, J.; Gutkind, J.S.; Coso, O.A. An Interplay between the P38 MAPK Pathway and AUBPs Regulates C-Fos MRNA Stability during Mitogenic Stimulation. Biochem. J. 2015, 467, 77–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Doller, A.; Schlepckow, K.; Schwalbe, H.; Pfeilschifter, J.; Eberhardt, W. Tandem Phosphorylation of Serines 221 and 318 by Protein Kinase Cdelta Coordinates MRNA Binding and Nucleocytoplasmic Shuttling of HuR. Mol. Cell. Biol. 2010, 30, 1397–1410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Fialcowitz-White, E.J.; Brewer, B.Y.; Ballin, J.D.; Willis, C.D.; Toth, E.A.; Wilson, G.M. Specific Protein Domains Mediate Cooperative Assembly of HuR Oligomers on AU-Rich MRNA-Destabilizing Sequences. J. Biol. Chem. 2007, 282, 20948–20959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Liwak-Muir, U.; Dobson, C.C.; Naing, T.; Wylie, Q.; Chehade, L.; Baird, S.D.; Chakraborty, P.K.; Holcik, M. ERK8 Is a Novel HuR Kinase That Regulates Tumour Suppressor PDCD4 through a MiR-21 Dependent Mechanism. Oncotarget 2016, 7, 1439–1450. [Google Scholar] [CrossRef] [Green Version]
  155. Bergalet, J.; Fawal, M.; Lopez, C.; Desjobert, C.; Lamant, L.; Delsol, G.; Morello, D.; Espinos, E. HuR-Mediated Control of C/EBPbeta MRNA Stability and Translation in ALK-Positive Anaplastic Large Cell Lymphomas. Mol. Cancer Res. 2011, 9, 485–496. [Google Scholar] [CrossRef] [Green Version]
  156. Gummadi, L.; Taylor, L.; Curthoys, N.P. Concurrent Binding and Modifications of AUF1 and HuR Mediate the PH-Responsive Stabilization of Phosphoenolpyruvate Carboxykinase MRNA in Kidney Cells. Am. J. Physiol. Renal Physiol. 2012, 303, F1545–F1554. [Google Scholar] [CrossRef] [Green Version]
  157. Dickson, A.M.; Anderson, J.R.; Barnhart, M.D.; Sokoloski, K.J.; Oko, L.; Opyrchal, M.; Galanis, E.; Wilusz, C.J.; Morrison, T.E.; Wilusz, J. Dephosphorylation of HuR Protein during Alphavirus Infection Is Associated with HuR Relocalization to the Cytoplasm. J. Biol. Chem. 2012, 287, 36229–36238. [Google Scholar] [CrossRef] [Green Version]
  158. Mazroui, R.; Di Marco, S.; Clair, E.; von Roretz, C.; Tenenbaum, S.A.; Keene, J.D.; Saleh, M.; Gallouzi, I.-E. Caspase-Mediated Cleavage of HuR in the Cytoplasm Contributes to Pp32/PHAP-I Regulation of Apoptosis. J. Cell Biol. 2008, 180, 113–127. [Google Scholar] [CrossRef]
  159. von Roretz, C.; Gallouzi, I.-E. Protein Kinase RNA/FADD/Caspase-8 Pathway Mediates the Proapoptotic Activity of the RNA-Binding Protein Human Antigen R (HuR). J. Biol. Chem. 2010, 285, 16806–16813. [Google Scholar] [CrossRef] [Green Version]
  160. Beauchamp, P.; Nassif, C.; Hillock, S.; van der Giessen, K.; von Roretz, C.; Jasmin, B.J.; Gallouzi, I.-E. The Cleavage of HuR Interferes with Its Transportin-2-Mediated Nuclear Import and Promotes Muscle Fiber Formation. Cell Death Differ. 2010, 17, 1588–1599. [Google Scholar] [CrossRef] [Green Version]
  161. Zhou, H.-L.; Geng, C.; Luo, G.; Lou, H. The P97-UBXD8 Complex Destabilizes MRNA by Promoting Release of Ubiquitinated HuR from MRNP. Genes Dev. 2013, 27, 1046–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Lucchesi, C.; Sheikh, M.S.; Huang, Y. Negative Regulation of RNA-Binding Protein HuR by Tumor-Suppressor ECRG2. Oncogene 2016, 35, 2565–2573. [Google Scholar] [CrossRef] [PubMed]
  163. Ke, Y.; Han, Y.; Guo, X.; Wen, J.; Wang, K.; Jiang, X.; Tian, X.; Ba, X.; Boldogh, I.; Zeng, X. PARP1 Promotes Gene Expression at the Post-Transcriptiona Level by Modulating the RNA-Binding Protein HuR. Nat. Commun. 2017, 8, 14632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ke, Y.; Lv, X.; Fu, X.; Zhang, J.; Bohio, A.A.; Zeng, X.; Hao, W.; Wang, R.; Boldogh, I.; Ba, X. Poly(ADP-Ribosyl)Ation Enhances HuR Oligomerization and Contributes to pro-Inflammatory Gene MRNA Stabilization. Cell. Mol. Life Sci. 2021, 78, 1817–1835. [Google Scholar] [CrossRef]
  165. Bibli, S.-I.; Hu, J.; Sigala, F.; Wittig, I.; Heidler, J.; Zukunft, S.; Tsilimigras, D.I.; Randriamboavonjy, V.; Wittig, J.; Kojonazarov, B.; et al. Cystathionine γ Lyase Sulfhydrates the RNA Binding Protein Human Antigen R to Preserve Endothelial Cell Function and Delay Atherogenesis. Circulation 2019, 139, 101–114. [Google Scholar] [CrossRef]
  166. Deka, K.; Saha, S. Heat Stress Induced Arginylation of HuR Promotes Alternative Polyadenylation of Hsp70.3 by Regulating HuR Stability and RNA Binding. Cell Death Differ. 2021, 28, 730–747. [Google Scholar] [CrossRef]
  167. Kristensen, L.S.; Hansen, T.B.; Venø, M.T.; Kjems, J. Circular RNAs in Cancer: Opportunities and Challenges in the Field. Oncogene 2017, 37, 555–565. [Google Scholar] [CrossRef] [Green Version]
  168. Abdelmohsen, K.; Panda, A.C.; Munk, R.; Grammatikakis, I.; Dudekula, D.B.; De, S.; Kim, J.; Noh, J.H.; Kim, K.M.; Martindale, J.L.; et al. Identification of HuR Target Circular RNAs Uncovers Suppression of PABPN1 Translation by CircPABPN1. RNA Biol. 2017, 14, 361–369. [Google Scholar] [CrossRef] [Green Version]
  169. David, P.S.; Tanveer, R.; Port, J.D. FRET-Detectable Interactions between the ARE Binding Proteins, HuR and P37AUF1. RNA 2007, 13, 1453–1468. [Google Scholar] [CrossRef] [Green Version]
  170. Ripin, N.; Boudet, J.; Duszczyk, M.M.; Hinniger, A.; Faller, M.; Krepl, M.; Gadi, A.; Schneider, R.J.; Šponer, J.; Meisner-Kober, N.C.; et al. Molecular Basis for AU-Rich Element Recognition and Dimerization by the HuR C-Terminal RRM. Proc. Natl. Acad. Sci. USA 2019, 116, 2935–2944. [Google Scholar] [CrossRef] [Green Version]
  171. Soller, M.; White, K. ELAV Multimerizes on Conserved AU4-6 Motifs Important for Ewg Splicing Regulation. Mol. Cell. Biol. 2005, 25, 7580–7591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Meisner, N.-C.; Hintersteiner, M.; Mueller, K.; Bauer, R.; Seifert, J.-M.; Naegeli, H.-U.; Ottl, J.; Oberer, L.; Guenat, C.; Moss, S.; et al. Identification and Mechanistic Characterization of Low-Molecular-Weight Inhibitors for HuR. Nat. Chem. Biol. 2007, 3, 508–515. [Google Scholar] [CrossRef] [PubMed]
  173. Filippova, N.; Yang, X.; Ananthan, S.; Sorochinsky, A.; Hackney, J.R.; Gentry, Z.; Bae, S.; King, P.; Nabors, L.B. Hu Antigen R (HuR) Multimerization Contributes to Glioma Disease Progression. J. Biol. Chem. 2017, 292, 16999–17010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Gantt, K.; Cherry, J.; Tenney, R.; Karschner, V.; Pekala, P.H. An Early Event in Adipogenesis, the Nuclear Selection of the CCAAT Enhancer-Binding Protein β (C/EBPβ) MRNA by HuR and Its Translocation to the Cytosol *. J. Biol. Chem. 2005, 280, 24768–24774. [Google Scholar] [CrossRef] [Green Version]
  175. Cherry, J.; Karschner, V.; Jones, H.; Pekala, P.H. HuR, an RNA-Binding Protein, Involved in the Control of Cellular Differentiation. In Vivo 2006, 20, 17–23. [Google Scholar]
  176. Siang, D.T.C.; Lim, Y.C.; Kyaw, A.M.M.; Win, K.N.; Chia, S.Y.; Degirmenci, U.; Hu, X.; Tan, B.C.; Walet, A.C.E.; Sun, L.; et al. The RNA-Binding Protein HuR Is a Negative Regulator in Adipogenesis. Nat. Commun. 2020, 11, 213. [Google Scholar] [CrossRef]
  177. Katsanou, V.; Papadaki, O.; Milatos, S.; Blackshear, P.J.; Anderson, P.; Kollias, G.; Kontoyiannis, D.L. HuR as a Negative Posttranscriptional Modulator in Inflammation. Mol. Cell 2005, 19, 777–789. [Google Scholar] [CrossRef]
  178. Gerbes, A.; Zoulim, F.; Tilg, H.; Dufour, J.-F.; Bruix, J.; Paradis, V.; Salem, R.; Peck-Radosavljevic, M.; Galle, P.R.; Greten, T.F.; et al. Gut Roundtable Meeting Paper: Selected Recent Advances in Hepatocellular Carcinoma. Gut 2018, 67, 380–388. [Google Scholar] [CrossRef]
  179. Villanueva, A. Hepatocellular Carcinoma. N. Engl. J. Med. 2019, 380, 1450–1462. [Google Scholar] [CrossRef] [Green Version]
  180. Rizzo, A.; Ricci, A.D.; Gadaleta-Caldarola, G.; Brandi, G. First-Line Immune Checkpoint Inhibitor-Based Combinations in Unresectable Hepatocellular Carcinoma: Current Management and Future Challenges. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 1245–1251. [Google Scholar] [CrossRef]
  181. Rizzo, A.; Ricci, A.D. PD-L1, TMB, and Other Potential Predictors of Response to Immunotherapy for Hepatocellular Carcinoma: How Can They Assist Drug Clinical Trials? Expert Opin. Investig. Drugs 2022, 31, 415–423. [Google Scholar] [CrossRef]
  182. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC Strategy for Prognosis Prediction and Treatment Recommendation: The 2022 Update. J. Hepatol. 2022, 76, 681–693. [Google Scholar] [CrossRef] [PubMed]
  183. Massarweh, N.N.; El-Serag, H.B. Epidemiology of Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. Cancer Control 2017, 24, 1073274817729245. [Google Scholar] [CrossRef] [PubMed]
  184. Zabron, A.; Edwards, R.J.; Khan, S.A. The Challenge of Cholangiocarcinoma: Dissecting the Molecular Mechanisms of an Insidious Cancer. Dis. Model. Mech. 2013, 6, 281–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Krenzien, F.; Nevermann, N.; Krombholz, A.; Benzing, C.; Haber, P.; Fehrenbach, U.; Lurje, G.; Pelzer, U.; Pratschke, J.; Schmelzle, M.; et al. Treatment of Intrahepatic Cholangiocarcinoma-A Multidisciplinary Approach. Cancers 2022, 14, 362. [Google Scholar] [CrossRef] [PubMed]
  186. Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next Horizon in Mechanisms and Management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef] [PubMed]
  187. Embade, N.; Fernández-Ramos, D.; Varela-Rey, M.; Beraza, N.; Sini, M.; de Juan, V.G.; Woodhoo, A.; Martínez-López, N.; Rodríguez-Iruretagoyena, B.; Bustamante, F.J.; et al. Mdm2 Regulates HuR Stability in Human Liver and Colon Cancer through Neddylation. Hepatology 2012, 55, 1237–1248. [Google Scholar] [CrossRef] [Green Version]
  188. Zhu, H.; Berkova, Z.; Mathur, R.; Sehgal, L.; Khashab, T.; Tao, R.-H.; Ao, X.; Feng, L.; Sabichi, A.L.; Blechacz, B.; et al. HuR Suppresses Fas Expression and Correlates with Patient Outcome in Liver Cancer. Mol. Cancer Res. 2015, 13, 809–818. [Google Scholar] [CrossRef] [Green Version]
  189. Vázquez–Chantada, M.; Fernández–Ramos, D.; Embade, N.; Martínez–Lopez, N.; Varela–Rey, M.; Woodhoo, A.; Luka, Z.; Wagner, C.; Anglim, P.P.; Finnell, R.H.; et al. HuR/Methyl-HuR and AUF1 Regulate the MAT Expressed During Liver Proliferation, Differentiation, and Carcinogenesis. Gastroenterology 2010, 138, 1943–1953.e3. [Google Scholar] [CrossRef] [Green Version]
  190. Ally, A.; Balasundaram, M.; Carlsen, R.; Chuah, E.; Clarke, A.; Dhalla, N.; Holt, R.A.; Jones, S.J.M.; Lee, D.; Ma, Y.; et al. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell 2017, 169, 1327–1341.e23. [Google Scholar] [CrossRef] [Green Version]
  191. Toyota, K.; Murakami, Y.; Kondo, N.; Uemura, K.; Nakagawa, N.; Takahashi, S.; Sueda, T. Cytoplasmic Hu-Antigen R (HuR) Expression Is Associated with Poor Survival in Patients with Surgically Resected Cholangiocarcinoma Treated with Adjuvant Gemcitabine-Based Chemotherapy. Ann. Surg. Oncol. 2018, 25, 1202–1210. [Google Scholar] [CrossRef] [PubMed]
  192. Villanueva, A.; Chiang, D.Y.; Newell, P.; Peix, J.; Thung, S.; Alsinet, C.; Tovar, V.; Roayaie, S.; Minguez, B.; Sole, M.; et al. Pivotal Role of MTOR Signaling in Hepatocellular Carcinoma. Gastroenterology 2008, 135, 1972–1983.e11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Wang, L.; Ding, Q.; Zhao, L.; Pan, Y.; Song, Z.; Qin, Y.; Yan, X. Decreased BMP-7 and p-Smad1/5/8 Expression, and Increased Levels of Gremlin in Hepatocellular Carcinoma. Oncol. Lett. 2018, 16, 2113–2118. [Google Scholar] [CrossRef]
  194. Hung, C.-M.; Huang, W.-C.; Pan, H.-L.; Chien, P.-H.; Lin, C.-W.; Chen, L.-C.; Chien, Y.-F.; Lin, C.-C.; Leow, K.-H.; Chen, W.-S.; et al. Hepatitis B Virus X Upregulates HuR Protein Level to Stabilize HER2 Expression in Hepatocellular Carcinoma Cells. Bio. Res. Int. 2014, 2014, 827415. [Google Scholar] [CrossRef] [PubMed]
  195. Martínez–Chantar, M.L.; Vázquez–Chantada, M.; Garnacho, M.; Latasa, M.U.; Varela–Rey, M.; Dotor, J.; Santamaria, M.; Martínez–Cruz, L.A.; Parada, L.A.; Lu, S.C.; et al. S–Adenosylmethionine Regulates Cytoplasmic HuR Via AMP–Activated Kinase. Gastroenterology 2006, 131, 223–232. [Google Scholar] [CrossRef]
  196. Lai, K.K.Y.; Kweon, S.-M.; Chi, F.; Hwang, E.; Kabe, Y.; Higashiyama, R.; Qin, L.; Yan, R.; Wu, R.P.; Lai, K.; et al. Stearoyl-CoA Desaturase Promotes Liver Fibrosis and Tumor Development in Mice via a Wnt Positive-Signaling Loop by Stabilization of Low-Density Lipoprotein-Receptor-Related Proteins 5 and 6. Gastroenterology 2017, 152, 1477–1491. [Google Scholar] [CrossRef] [Green Version]
  197. Chen, Y.; Peng, C.; Chen, J.; Chen, D.; Yang, B.; He, B.; Hu, W.; Zhang, Y.; Liu, H.; Dai, L.; et al. WTAP Facilitates Progression of Hepatocellular Carcinoma via M6A-HuR-Dependent Epigenetic Silencing of ETS1. Mol. Cancer 2019, 18, 127. [Google Scholar] [CrossRef] [Green Version]
  198. Zhang, R.; Wang, J. HuR Stabilizes TFAM MRNA in an ATM/P38-Dependent Manner in Ionizing Irradiated Cancer Cells. Cancer Sci. 2018, 109, 2446–2457. [Google Scholar] [CrossRef] [Green Version]
  199. Zhang, L.-F.; Lou, J.-T.; Lu, M.-H.; Gao, C.; Zhao, S.; Li, B.; Liang, S.; Li, Y.; Li, D.; Liu, M.-F. Suppression of MiR-199a Maturation by HuR Is Crucial for Hypoxia-Induced Glycolytic Switch in Hepatocellular Carcinoma. EMBO J. 2015, 34, 2671–2685. [Google Scholar] [CrossRef] [Green Version]
  200. Cao, C.; Sun, J.; Zhang, D.; Guo, X.; Xie, L.; Li, X.; Wu, D.; Liu, L. The Long Intergenic Noncoding RNA UFC1, a Target of MicroRNA 34a, Interacts with the MRNA Stabilizing Protein HuR to Increase Levels of β-Catenin in HCC Cells. Gastroenterology 2015, 148, 415–426.e18. [Google Scholar] [CrossRef]
  201. Huang, J.-F.; Jiang, H.-Y.; Cai, H.; Liu, Y.; Zhu, Y.-Q.; Lin, S.-S.; Hu, T.-T.; Wang, T.-T.; Yang, W.-J.; Xiao, B.; et al. Genome-Wide Screening Identifies Oncofetal LncRNA Ptn-Dt Promoting the Proliferation of Hepatocellular Carcinoma Cells by Regulating the Ptn Receptor. Oncogene 2019, 38, 3428–3445. [Google Scholar] [CrossRef] [PubMed]
  202. He, X.; Zheng, Y.; Zhang, Y.; Gan, Y.; Zhou, Y.; Liang, H.; Wu, D.; Ge, W.; Deng, J.; Xu, X. Long Non-Coding RNA AK058003, as a Precursor of MiR-15a, Interacts with HuR to Inhibit the Expression of γ-Synuclein in Hepatocellular Carcinoma Cells. Oncotarget 2017, 8, 9451–9465. [Google Scholar] [CrossRef] [PubMed]
  203. Liu, B.; Yang, G.; Wang, X.; Liu, J.; Lu, Z.; Wang, Q.; Xu, B.; Liu, Z.; Li, J. CircBACH1 (Hsa_circ_0061395) Promotes Hepatocellular Carcinoma Growth by Regulating P27 Repression via HuR. J. Cell. Physiol. 2020, 235, 6929–6941. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, H.; Lan, T.; Li, H.; Xu, L.; Chen, X.; Liao, H.; Chen, X.; Du, J.; Cai, Y.; Wang, J.; et al. Circular RNA CircDLC1 Inhibits MMP1-Mediated Liver Cancer Progression via Interaction with HuR. Theranostics 2021, 11, 1396–1411. [Google Scholar] [CrossRef]
  205. Wang, Y.; Gao, R.; Li, J.; Tang, S.; Li, S.; Tong, Q.; Li, S. Downregulation of Hsa_circ_0074854 Suppresses the Migration and Invasion in Hepatocellular Carcinoma via Interacting with HuR and via Suppressing Exosomes-Mediated Macrophage M2 Polarization. Int. J. Nanomed. 2021, 16, 2803–2818. [Google Scholar] [CrossRef]
  206. Martínez-López, N.; Varela-Rey, M.; Fernández-Ramos, D.; Woodhoo, A.; Vázquez-Chantada, M.; Embade, N.; Espinosa-Hevia, L.; Bustamante, F.J.; Parada, L.A.; Rodriguez, M.S.; et al. Activation of LKB1-Akt Pathway Independent of Phosphoinositide 3-Kinase Plays a Critical Role in the Proliferation of Hepatocellular Carcinoma from Nonalcoholic Steatohepatitis. Hepatology 2010, 52, 1621–1631. [Google Scholar] [CrossRef] [Green Version]
  207. Chen, T.; Dai, X.; Dai, J.; Ding, C.; Zhang, Z.; Lin, Z.; Hu, J.; Lu, M.; Wang, Z.; Qi, Y.; et al. AFP Promotes HCC Progression by Suppressing the HuR-Mediated Fas/FADD Apoptotic Pathway. Cell Death Dis. 2020, 11, 822. [Google Scholar] [CrossRef]
  208. Ji, E.; Kim, C.; Kang, H.; Ahn, S.; Jung, M.; Hong, Y.; Tak, H.; Lee, S.; Kim, W.; Lee, E.K. RNA Binding Protein HuR Promotes Autophagosome Formation by Regulating Expression of Autophagy-Related Proteins 5, 12, and 16 in Human Hepatocellular Carcinoma Cells. Mol. Cell. Biol. 2019, 39, e00508-18. [Google Scholar] [CrossRef] [Green Version]
  209. Papatheofani, V.; Levidou, G.; Sarantis, P.; Koustas, E.; Karamouzis, M.V.; Pergaris, A.; Kouraklis, G.; Theocharis, S. HuR Protein in Hepatocellular Carcinoma: Implications in Development, Prognosis and Treatment. Biomedicines 2021, 9, 119. [Google Scholar] [CrossRef]
  210. Mohibi, S.; Chen, X.; Zhang, J. Cancer the’RBP’eutics-RNA-Binding Proteins as Therapeutic Targets for Cancer. Pharmacol. Ther. 2019, 203, 107390. [Google Scholar] [CrossRef]
  211. Blanco, F.F.; Jimbo, M.; Wulfkuhle, J.; Gallagher, I.; Deng, J.; Enyenihi, L.; Meisner-Kober, N.; Londin, E.; Rigoutsos, I.; Sawicki, J.A.; et al. The MRNA-Binding Protein HuR Promotes Hypoxia-Induced Chemoresistance through Posttranscriptional Regulation of the Proto-Oncogene PIM1 in Pancreatic Cancer Cells. Oncogene 2016, 35, 2529–2541. [Google Scholar] [CrossRef]
  212. Romeo, C.; Weber, M.C.; Zarei, M.; DeCicco, D.; Chand, S.N.; Lobo, A.D.; Winter, J.M.; Sawicki, J.A.; Sachs, J.N.; Meisner-Kober, N.; et al. HuR Contributes to TRAIL Resistance by Restricting Death Receptor 4 Expression in Pancreatic Cancer Cells. Mol. Cancer Res. 2016, 14, 599–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Lang, M.; Berry, D.; Passecker, K.; Mesteri, I.; Bhuju, S.; Ebner, F.; Sedlyarov, V.; Evstatiev, R.; Dammann, K.; Loy, A.; et al. HuR Small-Molecule Inhibitor Elicits Differential Effects in Adenomatosis Polyposis and Colorectal Carcinogenesis. Cancer Res. 2017, 77, 2424–2438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. D’Agostino, V.G.; Lal, P.; Mantelli, B.; Tiedje, C.; Zucal, C.; Thongon, N.; Gaestel, M.; Latorre, E.; Marinelli, L.; Seneci, P.; et al. Dihydrotanshinone-I Interferes with the RNA-Binding Activity of HuR Affecting Its Post-Transcriptional Function. Sci. Rep. 2015, 5, 16478. [Google Scholar] [CrossRef]
  215. Chae, M.-J.; Sung, H.Y.; Kim, E.-H.; Lee, M.; Kwak, H.; Chae, C.H.; Kim, S.; Park, W.-Y. Chemical Inhibitors Destabilize HuR Binding to the AU-Rich Element of TNF-Alpha MRNA. Exp. Mol. Med. 2009, 41, 824–831. [Google Scholar] [CrossRef] [PubMed]
  216. Wu, X.; Lan, L.; Wilson, D.M.; Marquez, R.T.; Tsao, W.-C.; Gao, P.; Roy, A.; Turner, B.A.; McDonald, P.; Tunge, J.A.; et al. Identification and Validation of Novel Small Molecule Disruptors of HuR-MRNA Interaction. ACS Chem. Biol. 2015, 10, 1476–1484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Muralidharan, R.; Mehta, M.; Ahmed, R.; Roy, S.; Xu, L.; Aubé, J.; Chen, A.; Zhao, Y.D.; Herman, T.; Ramesh, R.; et al. HuR-Targeted Small Molecule Inhibitor Exhibits Cytotoxicity towards Human Lung Cancer Cells. Sci. Rep. 2017, 7, 9694. [Google Scholar] [CrossRef] [PubMed]
  218. Allegri, L.; Baldan, F.; Roy, S.; Aubé, J.; Russo, D.; Filetti, S.; Damante, G. The HuR CMLD-2 Inhibitor Exhibits Antitumor Effects via MAD2 Downregulation in Thyroid Cancer Cells. Sci. Rep. 2019, 9, 7374. [Google Scholar] [CrossRef]
  219. Wu, X.; Gardashova, G.; Lan, L.; Han, S.; Zhong, C.; Marquez, R.T.; Wei, L.; Wood, S.; Roy, S.; Gowthaman, R.; et al. Targeting the Interaction between RNA-Binding Protein HuR and FOXQ1 Suppresses Breast Cancer Invasion and Metastasis. Commun. Biol. 2020, 3, 193. [Google Scholar] [CrossRef]
  220. Yang, L.-Q.; Yu, S.-P.; Yang, Y.-T.; Zhao, Y.-S.; Wang, F.-Y.; Chen, Y.; Li, Q.-H.; Tian, P.; Zhu, Y.-Y.; Zhang, J.-G.; et al. Muscone Derivative ZM-32 Inhibits Breast Tumor Angiogenesis by Suppressing HuR-Mediated VEGF and MMP9 Expression. Biomed. Pharmacother. 2021, 136, 111265. [Google Scholar] [CrossRef]
  221. Yang, H.; Zheng, Y.; Li, T.W.H.; Peng, H.; Fernandez-Ramos, D.; Martínez-Chantar, M.L.; Rojas, A.L.; Mato, J.M.; Lu, S.C. Methionine Adenosyltransferase 2B, HuR, and Sirtuin 1 Protein Cross-Talk Impacts on the Effect of Resveratrol on Apoptosis and Growth in Liver Cancer Cells. J. Biol. Chem. 2013, 288, 23161–23170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Lee, J.-Y.; Chung, T.-W.; Choi, H.-J.; Lee, C.H.; Eun, J.S.; Han, Y.T.; Choi, J.-Y.; Kim, S.-Y.; Han, C.-W.; Jeong, H.-S.; et al. A Novel Cantharidin Analog N-Benzylcantharidinamide Reduces the Expression of MMP-9 and Invasive Potentials of Hep3B via Inhibiting Cytosolic Translocation of HuR. Biochem. Biophys. Res. Commun. 2014, 447, 371–377. [Google Scholar] [CrossRef] [PubMed]
  223. Doller, A.; Badawi, A.; Schmid, T.; Brauss, T.; Pleli, T.; zu Heringdorf, D.M.; Piiper, A.; Pfeilschifter, J.; Eberhardt, W. The Cytoskeletal Inhibitors Latrunculin A and Blebbistatin Exert Antitumorigenic Properties in Human Hepatocellular Carcinoma Cells by Interfering with Intracellular HuR Trafficking. Exp. Cell Res. 2015, 330, 66–80. [Google Scholar] [CrossRef] [PubMed]
  224. Barbier-torres, L.; Fernández-ramos, D.; Martínez-chantar, M.L. The Levels of the RNA Binding Protein Hu Antigen R Determine the Druggability of the Neddylation Pathway in Liver Cancer. RNA Dis. 2016, 3, 1–10. [Google Scholar] [CrossRef] [Green Version]
  225. Sanna, M.D.; Quattrone, A.; Galeotti, N. Silencing of the RNA-Binding Protein HuR Attenuates Hyperalgesia and Motor Disability in Experimental Autoimmune Encephalomyelitis. Neuropharmacology 2017, 123, 116–125. [Google Scholar] [CrossRef]
  226. Muralidharan, R.; Babu, A.; Amreddy, N.; Basalingappa, K.; Mehta, M.; Chen, A.; Zhao, Y.D.; Kompella, U.B.; Munshi, A.; Ramesh, R. Folate Receptor-Targeted Nanoparticle Delivery of HuR-RNAi Suppresses Lung Cancer Cell Proliferation and Migration. J. Nanobiotechnol. 2016, 14, 47. [Google Scholar] [CrossRef] [Green Version]
  227. Katsanou, V.; Milatos, S.; Yiakouvaki, A.; Sgantzis, N.; Kotsoni, A.; Alexiou, M.; Harokopos, V.; Aidinis, V.; Hemberger, M.; Kontoyiannis, D.L. The RNA-Binding Protein Elavl1/HuR Is Essential for Placental Branching Morphogenesis and Embryonic Development. Mol. Cell. Biol. 2009, 29, 2762–2776. [Google Scholar] [CrossRef] [Green Version]
  228. Sun, D.-Q.; Wang, Y.; Liu, D.-G. Cancer Cell Growth Suppression by a 62nt AU-Rich RNA from C/EBPβ 3’UTR through Competitive Binding with HuR. Biochem. Biophys. Res. Commun. 2012, 426, 122–128. [Google Scholar] [CrossRef]
  229. Nimjee, S.M.; White, R.R.; Becker, R.C.; Sullenger, B.A. Aptamers as Therapeutics. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 61–79. [Google Scholar] [CrossRef]
Figure 1. HuR domain organization, structures, and sequence alignment. (A) Schematic domain organization of human HuR. Black numbers indicate the residues flanking each domain. RNPs and residues flanking RNPs are colored in cyan. C13 and W261, implicated in dimerization, are highlighted in yellow. Susceptible lysines to become NEDDylated are highlighted in purple. N and C indicate N-terminal and C-terminal endings, respectively. (B) Ribbon structures of human HuR RRM domains. Side-chains of residues at RNPs are colored in cyan, those of C13 and W261 are highlighted in yellow; NEDDylated lysines are shown in purple. β-strands and α-helixes numbers are indicated. Left panel: NMR structure of RRM1 domain. Ten models are shown (PDB: 5SZW) [14]. Middle panel: X-ray structure of RRM12 tandem of HuR (PDB: 4ED5) [5]. Right panel: X-ray structure of RRM3 domain, in its dimeric form (PDB: 6GD1) [15]. (C) Sequence alignment of HuR, with homologous RNA binding proteins (ELAV, ELAVL-1, ELAVL-2, ELAVL-3, ELAVL-4, and PABP-4), from human and related organisms. Residues belonging to each RRM domain are shaded in grey. RNPs residues from HuR are highlighted in cyan, whereas C13 and W261 are in yellow. K283, K313, and K326 are highlighted in purple. Blue arrows represent β-strands, and red rectangles represent α-helixes of HuR.
Figure 1. HuR domain organization, structures, and sequence alignment. (A) Schematic domain organization of human HuR. Black numbers indicate the residues flanking each domain. RNPs and residues flanking RNPs are colored in cyan. C13 and W261, implicated in dimerization, are highlighted in yellow. Susceptible lysines to become NEDDylated are highlighted in purple. N and C indicate N-terminal and C-terminal endings, respectively. (B) Ribbon structures of human HuR RRM domains. Side-chains of residues at RNPs are colored in cyan, those of C13 and W261 are highlighted in yellow; NEDDylated lysines are shown in purple. β-strands and α-helixes numbers are indicated. Left panel: NMR structure of RRM1 domain. Ten models are shown (PDB: 5SZW) [14]. Middle panel: X-ray structure of RRM12 tandem of HuR (PDB: 4ED5) [5]. Right panel: X-ray structure of RRM3 domain, in its dimeric form (PDB: 6GD1) [15]. (C) Sequence alignment of HuR, with homologous RNA binding proteins (ELAV, ELAVL-1, ELAVL-2, ELAVL-3, ELAVL-4, and PABP-4), from human and related organisms. Residues belonging to each RRM domain are shaded in grey. RNPs residues from HuR are highlighted in cyan, whereas C13 and W261 are in yellow. K283, K313, and K326 are highlighted in purple. Blue arrows represent β-strands, and red rectangles represent α-helixes of HuR.
Cancers 14 02666 g001
Figure 2. Main regulatory axes involving HuR, which are associated with cell proliferation, invasion, metastasis, apoptosis, and autophagy during HCC. These include the transcriptional, post-transcriptional, and post-translational modulators of HuR function, in addition to HuR target transcripts.
Figure 2. Main regulatory axes involving HuR, which are associated with cell proliferation, invasion, metastasis, apoptosis, and autophagy during HCC. These include the transcriptional, post-transcriptional, and post-translational modulators of HuR function, in addition to HuR target transcripts.
Cancers 14 02666 g002
Table 1. Small-molecule inhibitors controlling HuR function, tested in hepatobiliary tumors.
Table 1. Small-molecule inhibitors controlling HuR function, tested in hepatobiliary tumors.
MoleculeTypeEffectRef.
ZM-32Synthetic
muscone-derivative
Inhibition of HuR binding to target mRNAs[220]
Resveratrol (RSV)Naturally occurringIncrease in HuR mRNA and
protein expression
[221]
N-BenzylcantharidinamideSynthetic analogue of the naturally
occurring
cantharidine drug
Inhibition of HuR translocation to the cytosol[222]
Latrunculin ANaturally occurringInhibition of HuR translocation to the cytosol[223]
BlebbistatinNaturally occurringInhibition of HuR translocation to the cytosol[223]
PevonedistatSynthetic
NEDDylation
inhibitor
HuR destabilization[224]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lachiondo-Ortega, S.; Delgado, T.C.; Baños-Jaime, B.; Velázquez-Cruz, A.; Díaz-Moreno, I.; Martínez-Chantar, M.L. Hu Antigen R (HuR) Protein Structure, Function and Regulation in Hepatobiliary Tumors. Cancers 2022, 14, 2666. https://doi.org/10.3390/cancers14112666

AMA Style

Lachiondo-Ortega S, Delgado TC, Baños-Jaime B, Velázquez-Cruz A, Díaz-Moreno I, Martínez-Chantar ML. Hu Antigen R (HuR) Protein Structure, Function and Regulation in Hepatobiliary Tumors. Cancers. 2022; 14(11):2666. https://doi.org/10.3390/cancers14112666

Chicago/Turabian Style

Lachiondo-Ortega, Sofia, Teresa Cardoso Delgado, Blanca Baños-Jaime, Alejandro Velázquez-Cruz, Irene Díaz-Moreno, and María Luz Martínez-Chantar. 2022. "Hu Antigen R (HuR) Protein Structure, Function and Regulation in Hepatobiliary Tumors" Cancers 14, no. 11: 2666. https://doi.org/10.3390/cancers14112666

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop