Abstract
Glioblastoma (GBM) is a malignant and aggressive brain tumor with a median survival of ~ 15 months. Resistance to treatment arises from the extensive cellular and molecular heterogeneity in the three major components: glioma tumor cells, glioma stem cells, and tumor-associated microglia and macrophages. Within this triad, there is a complex network of intrinsic and secreted factors that promote classic hallmarks of cancer, including angiogenesis, resisting cell death, proliferation, and immune evasion. A regulatory node connecting these diverse pathways is at the posttranscriptional level as mRNAs encoding many of the key drivers contain adenine- and uridine rich elements (ARE) in the 3’ untranslated region. Human antigen R (HuR) binds to ARE-bearing mRNAs and is a major positive regulator at this level. This review focuses on basic concepts of ARE-mediated RNA regulation and how targeting HuR with small molecule inhibitors represents a plausible strategy for a multi-pronged therapeutic attack on GBM.
Keywords: posttranscriptional regulation, GBM, Adenine- and Uridine-rich elements, cytoplasmic translocation, HuR multimerization, small molecule inhibitors
Graphical Abstract

1. Introduction
Glioblastoma (GBM) is one of the most aggressive and invasive cancers of central nervous system (CNS) and comprises about 50% of all malignant brain tumors in adults [1]. Despite many clinical trials of multimodal therapeutic interventions, the prognosis of GBM remains poor with less than 5% of patients surviving beyond 5 years, a statistic that has not changed over 3 decades [2]. Major obstacles to the development of effective treatment arise from the incredible adaptability of the tumor to thrive under adverse conditions. Resistance to therapy arises from a small population of cells within GBM called glioma stem cells (GSC) which are defined by their ability to self-renew and generate heterogeneous tumors after transplantation [3, 4]. GSCs can thrive in harsh conditions such as hypoxia and acidosis, resist apoptosis and maintain tumorigenicity. The malignant phenotype is further supported by a favorable tumor microenvironment (TME). The TME is shaped by a complex glioma cell secretome that draws in tumor-associated microglia/macrophages (TAMs) and other immune cells to support proliferation, survival, parenchymal infiltration, and evasion of anti-tumor immune responses [5, 6]. Up to 40% of the GBM tumor mass is comprised of TAMs [5]. Effective therapies therefore need to target the multiple cellular and molecular components in GBM. A common regulatory thread for many of the intracellular and secreted factors, including cytokines, chemokines, anti-apoptotic factors, growth factors, and cell cycle regulators, that drive these tumor-promoting pathways is at the posttranscriptional level [7]. The mRNAs encoding these factors contain adenine- and/or uridine-rich elements (classically and historically referred to as the ARE) in the 3’ untranslated region (UTR) that govern RNA stability and translational efficiency [8–11]. These dual levels of regulation enable rapid changes in protein expression programs to maintain homeostasis and viability following cell damage [12]. Prime triggers of cell damage/cellular stress in the TME include hypoxia, acidosis, and glucose deficiency [3, 13]. A key factor in the posttranscriptional adaptive response of GBM to resist treatment and sustain growth under adverse conditions is the ARE binding protein (ARE-BP) HuR. In this review, we will describe the basic elements of ARE-mediated posttranscriptional regulation, the relevant mRNA targets, and how targeting HuR represents a plausible strategy for a multi-pronged therapeutic attack on GBM.
2. ARE-mediated posttranscriptional regulation
The field of ARE-mediated RNA regulation emerged more than three decades ago when Shaw and Kamen first observed that the normally stable β-globin mRNA transcript underwent rapid degradation simply by inserting an ARE into its 3’ UTR [11]. The ARE was historically defined by the presence of a core pentameric AUUUA motif, sometimes expanded to the nonamer UUAUUUAUU [14], but many other U- and AU-rich sequences, as well as sequences rich in other nucleotides, have been found to confer altered mRNA stability [15–17]. AREs are found in noncoding portions of mRNAs (predominately 3’ UTRs) in up to 85% of human genes, and in introns of more than 50% of human genes [18, 19]. A list of GBM-relevant mRNAs that are regulated by AREs is shown in Table 1. From the scope of function of the encoded proteins, one can appreciate the potentially broad impact that ARE-mediated regulation has on the biology of this tumor and other cancers. Through an interplay with trans-acting factors, including ARE-BPs and ncRNAs (miRNAs and lncRNAs), the ARE modulates two connected but distinct levels of gene regulation, RNA stability and translational efficiency (Fig. 1) [9, 20, 21]. RNA stability affects the overall levels of mRNA present in the cytoplasm, whereas translational efficiency reflects the actual utilization of mRNA for protein synthesis.
Table 1.
HuR-regulated mRNAs in glioblastoma.
| ARE Target | Role in Glioblastoma | Posttranscriptional Regulation | References | |||
|---|---|---|---|---|---|---|
| Glioma Cell | GSC | TME | ↑ mRNA Stability | ↓ mRNA Stability | ||
| Arg1 | ↑ TAM Recruitment ↑ Immune suppression |
X | [108, 128] | |||
| Bcl2 | ↓ Apoptosis | ↓ Apoptosis | X | [94, 97, 129, 130] | ||
| Bcl-XL | ↓ Apoptosis ↑ Migration ↑ Invasion |
↑ Proliferation ↑ Angiogenesis |
X | [94, 97, 131] | ||
| CCL2 | ↑ TAM Recruitment ↑ Immune Suppression ↑ Angiogenesis |
X | [132–135] | |||
| CCL5 | ↑ Migration ↑ Invasion |
↑ TAM Recruitment | X | [108, 136, 137] | ||
| CCNB2 | ↑ Proliferation | X | [54, 138] | |||
| CCND1 | ↑ Proliferation | X | [54, 139] | |||
| CCNE1 | ↑ Proliferation ↓ Apoptosis |
X | [54, 140] | |||
| c-FOS | ↑ Proliferation ↓ Apoptosis |
X | [54, 141] | |||
| cIAP2 | ↓ Apoptosis | ↑ Self-Renewal ↓ Apoptosis |
X | [97, 142] | ||
| c-Myc | ↑ Proliferation | ↑ Proliferation ↑ Self-Renewal |
X | [54, 143, 144] | ||
| COX2 | ↑ Proliferation | ↑ Immune Suppression ↑ Angiogenesis |
X | [86, 145–147] | ||
| CXCL1 | ↑ Proliferation | ↑ TAM Recruitment ↑ Immune Suppression |
X | [108, 135, 148, 149] | ||
| CXCL2 | ↓ Immune Destruction ↑ TAM Recruitment |
X | [108, 149] | |||
| CXCL3 | ↓ Immune Destruction | X | [149, 150] | |||
| CXCL8 (IL-8) | ↑ Proliferation | ↑ Proliferation | ↑ TAM Recruitment | X | [94, 97, 151–153] | |
| ↑ Migration ↑ Invasion |
↑ Self-Renewal | |||||
| CXCL1 0 | ↑ DNA Replication ↑ Proliferation |
↑ TAM Recruitment ↑ Immune Suppression |
X | [108, 154] | ||
| CXCR4 | ↑ Invasion ↑ Metastasis |
↑ Self-Renewal | ↑ Angiogenesis | X | [155–157] | |
| DR5 | ↑ Apoptosis | X | [97, 158] | |||
| EGF | ↑ Proliferation ↑ Metastasis |
↑ Angiogenesis | X | [54, 159, 160] | ||
| GM-CSF | ↑ Proliferation | ↑ Proliferation | ↑ TAM Recruitment ↑ Angiogenesis |
X | [54, 161–163] | |
| HIF1α | ↑ Invasion | ↑ Proliferation ↑ Self-Renewal |
↑ Angiogenesis | [97, 164] | ||
| IL-1β | ↑ Proliferation | ↑ Proliferation ↑ Self-Renewal |
X | [97, 108, 135, 165] | ||
| IL-4 | ↑ Immune Destruction ↓ Apoptosis |
X | [166, 167] | |||
| IL-6 | ↑ Migration ↑ Invasion |
↑ Migration ↑ Invasion ↑ Self-Renewal |
X | [97, 108, 135] | ||
| IL-10 | ↑ Proliferation ↑ Migration ↑ Invasion |
↑ Immune Suppression | X | [108, 168] | ||
| IL-13 | ↑ Apoptosis | ↑ Apoptosis | X | [169, 170] | ||
| iNOS | ↑ Migration ↑ Invasion |
↑ Migration ↑ Invasion ↑ Self-Renewal |
↑ Immune Suppression | X | [97, 171] | |
| Mcl-1 | ↓ Apoptosis | X | [94, 172] | |||
| MMP-2 | ↑ Invasion | X | [108] | |||
| MMP-9 | ↑ Invasion | X | [108, 173] | |||
| MMP12 | ↑ Invasion | X | [135, 149] | |||
| PDCD4 | ↓ Proliferation ↑ Apoptosis |
X | [174–176] | |||
| PD-L1 | ↑ Immune Suppression | X | [97, 108, 116, 177] | |||
| TGF-β1 | ↑ Immune Suppression ↑ Angiogenesis |
X | [97, 108, 178] | |||
| TGF-β2 | ↑ Proliferation ↑ Self-Renewal |
↑ Immune Suppression ↑ Angiogenesis |
X | [97, 178] | ||
| TNF-α | ↑ Migration ↑ Invasion |
↑ Migration ↑ Invasion | X | [97, 108, 135] | ||
| VEGF-A | ↑ Angiogenesis | X | [97, 108, 179] | |||
Abbreviations
ARE, AU and/or U-rich element; GSC, glioma stem cell; HuR, Human Antigen R; TAM, Tumor-associated macrophages/microglia; TME, Tumor microenvironment
Figure 1: General principles of ARE-mediated posttranscriptional regulation and the role of HuR.

In the nucleus, HuR binds to ARE regions of some newly transcribed pre-mRNAs and participates in splicing and mRNA maturation. HuR can help translocate to the cytoplasm some ARE-bearing mRNAs, and generally promotes the stabilization and translation of bound mRNAs. Factors that promote mRNA decay, which include microRNAs and ARE-binding proteins TTP, KSRP and AUF1, are blocked from binding to the mRNA. If HuR is retained in the nucleus, has impaired RNA binding, or is sequestered by non-coding RNAs, RNA destabilizers and/or the microRNA/RISC (miRISC) complex gain access to the mRNA which is then transported to exosomes or P bodies for degradation.
3. The discovery of HuR and the link to AREs
HuR (also referred to as ELAVL1 or HuA) belongs to the Embryonic-Lethal Abnormal Vision (ELAV) family of RNA-binding proteins and was cloned based on sequence homology to the neuron-specific homologue, HuD, which was first identified as a target of anti-Hu paraneoplastic autoantibodies in patients with small cell lung cancer [22–25]. The basic structure of the ELAV family, which also includes neuronal members Hel-N1 (HuB or ELAVL2) and HuC (ELAV3), is characterized by the presence of three RNA-recognition motifs (RRMs), a hinge region between RRMs 2 and 3, and a unique N-terminal region (Fig. 2) [23, 26]. Sequence homology between HuR and neuronal ELAV members is high in the RRMs (70–90%) but drops considerably in the other regions (~15–40%) and this difference is underscored by the significantly lower immunoreactivity of HuR to anti-Hu autoantibodies [26, 27]. The broader expression pattern of HuR, beyond neural tissues and small cell lung cancer, was initially established by RNA studies [22, 25]. The link between the ELAV family and ARE binding was first made with Hel-N1 using a random RNA selection procedure which identified a binding predilection for U-rich sequences flanked by adenines and cytidines (C) [28]. RNA-binding assays then confirmed that Hel-N1 could bind to AREs in the 3’ UTRs of c-Myc, c-Fos and ID1 [26, 28]. HuR was found to have RNA binding preferences, including C- and U-rich sequences [22, 29]. Subsequent transcriptome-wide binding analyses with photoactivatable ribonucleoside enhanced crosslinking and immunoprecipitation (PAR-CLIP) with HuR confirmed these initial binding preferences and identified multiple HuR binding sites outside of the 3’ UTR including intronic sequences and 5’ UTRs [15, 30].
Figure 2: Structural features of HuR.

Schematic domain organization of HuR. The N-terminal RRM and central RRM 2 domains are separated by a short linker, while C-terminal RRM3 is connected to the tandem domains by a long intrinsically disordered hinge region (upper panel). Each of the RRMs has a cognate topology of β1α1β2β3α2β4, where the antiparallel β sheets are arranged against 2 α helices that are perpendicularly oriented (lower left panel). The N-terminal cysteine (cys13) residue, and/or C-terminal tryptophan residues (trp261, trp261 and trp271) are essential for multimerization of HuR which fine-tunes its function (lower right panel) [32, 36–38].
Detailed analyses of HuR-RNA interactions show that the N-terminal RRM1 and central RRM2 domains are essential for ARE recognition, whereas the C-terminal RRM3 interacts with poly-A and short U-rich sequences of target mRNAs [31–33]. ARE binding with RRM1 induces a conformational change in HuR, allowing further interactions with the inter-domain linker and RRM2 which enhances the ARE-binding affinity of HuR [34]. However, a recent report suggests RNA recognition involves all three RRMs of the full-length HuR [33]. Nevertheless, RRM3 discriminates between various ARE motifs and preferably binds to poly-U stretches [32, 35]. RRM3 also acts as multifunctional domain, helping in HuR multimerization and binding RNA targets using two different surfaces on opposite sides of the domain [32, 35]. In addition, multimerization was shown to be dependent on the N-terminal cysteine (cys13) residue, and/or C-terminal tryptophan residues (trp261, trp261 and trp271) of RRM3. Multimerization is pivotal for fine-tuning HuR function (Fig. 2) [32, 36–38]. The intrinsically disordered hinge region is also important for multimerization and RNA-binding activity [38–40]. The hinge and RRM3 were found to regulate cooperative assembly of HuR oligomers on the AREs [40]. In the same study, multimerization of HuR was shown to be dependent on the density of AREs in the target transcripts [40]. Further, a HuR nucleocytoplasmic shuttling sequence (HNS) resides in the hinge region which facilitates transport of HuR back to the nucleus [41].
4. The role of HuR in ARE-mediated RNA regulation
Although HuR is predominantly nuclear, its nuclear function is not well understood. It has been found to bind to and modulate splicing of some pre-mRNAs [42] and promote cytoplasmic export of some mature mRNAs (Fig. 1). In the cytoplasm, HuR remains bound to the mature mRNA and generally protects ARE mRNAs from degradation and promotes translation by facilitating polysomal association [9, 21, 43–45]. This shift is often in response to cellular stresses or mitogens [42]. Other ARE-BPs such as Tristetraprolin (TTP) and KH-like splicing protein (KSRP) compete for binding to the same AREs but instead promote RNA degradation by relocating the mRNA to exonucleases located within P bodies or exosomes in the cytoplasm [20, 44, 46–49]. In other cases, HuR prevents degradation and promotes translation by preventing the repressive function of microRNAs and the RNA-induced silencing complex (RISC) [50]. Thus, the state of ARE mRNA stability and translational efficiency reflects a balance between negative and positive ARE-BPs and miRNAs available for binding. In GBM, the equilibrium shifts toward HuR due to its high overall expression and increased cytoplasmic localization [38, 51]. The latter is considered necessary for HuR-driven oncogenesis and is in part driven by p38 MAPK, a kinase that is active in GBM and correlates with high tumor grade, invasion, and poor survival [52–54]. Phosphorylation by this kinase can enhance HuR translocation to the cytoplasm [55–57]. The equilibrium is further shifted toward mRNA stabilization as p38 MAPK-driven phosphorylation of TTP reduces its availability and functionality as a destabilizer [38, 44, 51, 55, 58–60]. Based on our analysis of primary GBM tissue and glioma tumor cells (GTC), TTP is extensively phosphorylated and p38 MAPK plays a major role in that modification [59]. With ectopic expression of TTP in GTCs, the equilibrium could be pushed toward destabilization of transcripts including VEGF and IL-8 mRNAs resulting in downregulation of VEGF and IL-8, attenuated proliferation and increased apoptosis [59]. Mutation of key serines to alanines prevented TTP phosphorylation, retained TTP activity and increased the susceptibility of GTCs to temozolomide [61]. Therapeutic strategies to target HuR will be discussed later in this review, but center around shifting the equilibrium toward RNA destabilization by blocking HuR’s ability to bind AREs and/or translocate to the cytoplasm.
5. Functional interplay between HuR and microRNAs
In addition to phosphorylation, subcellular localization, and interaction with other RNA-binding proteins, HuR function is richly controlled through microRNAs. This control is elicited in two main ways: through microRNA-mediated changes in HuR abundance in the cell, and through cooperative and competitive actions of HuR and microRNAs to modulate expression of other proteins (Fig. 1).
Several prominent microRNAs control HuR abundance by binding the ELAVL1 mRNA (which encodes HuR) in response to different stimuli [62–66] and modulate HuR production post-transcriptionally. On the one hand, miR-519-, miR-125a-, miR-16- and miR-34a-mediated repression of HuR synthesis reduces cancer cell proliferation and chemotherapeutic resistance and enhances apoptosis, thereby suppressing the oncogenic function of HuR [62, 64–66]. On the other hand, miR-125b-mediated repression of HuR synthesis has been associated with pro-oncogenic phenotypes, supporting a tumor suppressor function of HuR in this paradigm [63], while promotion of HuR translation by miR-155–5p was reported to support colon cancer metastasis (Al-Haidari et al., 2018). In addition to the regulation of HuR synthesis by binding ELAVL1/HuR mRNA, signal-dependent posttranslational mechanisms of protein degradation, such as ubiquitination and caspase-mediated proteolytic cleavage, can also control HuR abundance [63, 67–73].
Besides being regulated by microRNAs, HuR can also regulate the function of several microRNAs either competitively or cooperatively. The competitive regulation of microRNA function by HuR primarily includes examples in which HuR binding to the 3’UTR “hides” a microRNA-binding site and prevents it from being recognized by the miRISC [74]. For example, under amino acid deprivation, HuR translocates from nucleus to cytoplasm and derepresses the translation of CAT1 mRNA by antagonizing the action of miR-122 in hepatic carcinoma cells [50]. HuR also translocates in response to UVC irradiation and inhibits miR-125b-mediated translation repression of p53 mRNA by dissociating miRISC from the p53 mRNA 3’UTR in breast carcinoma cells [75]. A similar mechanism has been observed between HuR and different miRNAs such as miR-548c, miR-494, miR-16, and miR-200b in response to different stimuli (reviewed by Srikantan et al. [76]) (Fig. 1). In addition to a competitive function, HuR has been found to cooperate with miRNAs. For instance, HuR represses the expression of c-myc at a posttranscriptional level by recruiting let-7-loaded RISC to the 3’UTR of c-myc mRNA [77]. Further evidence suggests that HuR can also directly interact with miRNAs and regulate their function. For example, HuR interacts with miR-16, miR-29 and miR-1192, and thereby derepresses the translation of COX-2, A20 and HMGB1 mRNAs respectively [78–80]. HuR also translocates from nucleus to cytoplasm upon LPS stimulation and sequesters miR-21 as “microRNA sponge”, thereby inhibiting miR-21-mediated translational suppression of PDCD4 mRNA [81] (Fig. 1). Finally, in yet another example of HuR-mediated microRNA regulation, HuR binding to miR-122 was implicated in the export of miR-122 into extracellular vesicles under conditions of metabolic stress [82].
6. HuR and the link to GBM
The potential relevance of HuR to cancer was surmised early on as ELAV family members bound mRNAs encoding proto-oncogenes including c-Myc, c-Fos, and GM-CSF [28]. One of the first ties to cancer was in GBM, where HuR was found to be overexpressed, particularly in peri-necrotic regions within the tumor [51]. These regions represent hypoxic zones and have marked upregulation of angiogenic proteins including COX-2, VEGF, and IL-8, all of which are encoded by ARE-bearing mRNAs [83–85]. This observation was underscored by Dixon et al. in colon cancer where HuR was also found to be overexpressed and linked to increased expression of oncogenic COX-2 through mRNA stabilization of its ARE-bearing transcript [86]. Since the original study of HuR and GBM, more extensive analyses of malignant glioma and normal brain tissue samples (TCGA and Gtex) have confirmed that HuR levels are significantly elevated and increase with tumor grade (Fig. 3A). Overall survival is also markedly worse (hazard ratio of 3.8) in patients with malignant gliomas having high HuR expression (Fig. 3B). The role of HuR in promoting malignant features of cancer and its correlation with worse clinical outcome extends to multiple extracranial cancers including breast, lung and pancreatic (reviewed in [87]).
Figure 3: HuR is upregulated in malignant glioma and correlates with poor overall survival.

(A) Comparison of HuR mRNA levels in GBM and lower grade (LG) glioma tissue with normal control brain tissue. The sample numbers are shown in parentheses. * P < 10–9. (B) Overall survival in patients with malignant glioma (GBM and LG gliomas) with high HuR mRNA levels (upper 50%, n = 338) compared to those with low HuR mRNA levels (lower 50%, n = 338). Hazard ratio for the high group is 3.8. 95% CI are shown. Data obtained from GEPIA (Gene Expression Profiling Interactive Analysis) (cancer-pku.cn).
The oncogenic role of HuR in GBM can best be understood by the relevance of the mRNA targets it regulates within the three major components of the tumor: GTCs, GSCs, and TAMs (Fig. 4 and Table 1). In this triad, there is a complex network of secreted and intrinsic factors regulated by HuR that promotes classic hallmarks of cancer, including angiogenesis, resisting cell death (survival), invasion, proliferation, genome instability and immune evasion [88]. In the initial studies linking HuR to GBM, in vitro RNA binding assays indicated that HuR bound with high affinity to ARE-rich 3’ UTRs of mRNAs encoding growth factors that support these cancer hallmarks including VEGF, IL-6, IL-8, and TGF-β [51, 89–92]. The positive effect of HuR on the expression of these and other oncogenic targets such as the Bcl-2 family of anti-apoptotic factors (Bcl-2, Mcl-1, and Bcl-XL) and the mTORC2 subunit Rictor, was demonstrated in GTCs [93–95]. Mechanisms for this positive effect include enhanced RNA stabilization and/or translation efficiency. The number of oncogenic mRNA targets negatively impacted in GTCs by HuR inhibition has expanded recently with the advent of small molecule inhibitors and has provided additional insight into the role of HuR in promoting tumor progression [96, 97]. These include immune evasion (PD-L1, iNOS, TGF-β1 and 2), angiogenesis (HIF-1α, COX-2), survival (cIAP2, XIAP) and GSC maintenance (HIF-1α, IL-6, TGF-β1 and 2, c-Myc), invasion (MMP-2, and 9, CCL5, ITG-A4), proliferation (Rictor, GM-CSF, CCNA1, CCNB2, CCND1, CCNE1). Interestingly, HuR associates with centromeres in GTCs driven by cyclin-dependent kinase 5 (CDK5)-mediated phosphorylation of HuR at a proline-directed serine 202. This leads to dissociation of CCNA1 (cyclin A) mRNA and its reduced translation, and defects in centromere duplication and cell cycle progression [98]. The ultimate impact on GTCs is promotion of mitotic abnormalities, generation of aneuploid cells, and centrosome amplification with resultant chromosomal instability, another hallmark of cancer and tumor heterogeneity.
Figure 4: HuR positively regulates ARE mRNAs in GSCs, GTCs and TAMs to drive tumor progression.

(A) Immunohistochemistry of biopsies obtained from the edge and core of a GBM tumor showing extensive HuR immunostaining in tumor cells and Iba1+ TAMs both in the tumor (Tu) and the surrounding brain parenchyma (Br). There is extensive translocation of HuR in Iba1+ cells as indicated by a merged signal with HuR (yellow). Dashed line indicates approximate border between Br and Tu. Scale bars, 100 μm. (B) HuR functions within each of the three major cellular components of GBM to promote intrinsic and secreted factors that drive GBM progression. GSCs, glioma stem cells; GTC,s glioma tumor cell; TAMs, tumor-associated microglia and macrophages; CTLs, Cytotoxic T lymphocytes; PMN-MDSCs, polymorphonuclear myeloid-derived suppressor cells;
GSCs represent the second component of the GBM triad supported by HuR. This small population of cells which drives treatment resistance and GBM progression can be enriched in cell culture through selection with the GSC marker, CD133 [99, 100]. Using three patient-derived GBM xenografts, we found that CD133+ cells were especially sensitive to HuR inhibition with viability assays indicating a ~ 2- to 10-fold lower IC50 than the parent cell line after treatment with the HuR inhibitor, MS-444 [45, 97]. Sublethal doses of MS-444 completely inhibited neurosphere formation, a hallmark of stemness. HuR inhibition also attenuated invasive properties of GSCs in culture. Since GSCs are major drivers of invasiveness [101], this may partially explain prior work showing a significantly reduced infiltration of GBM tumors intracranially when HuR was silenced [94]. The increased sensitivity of GSCs to HuR inhibition may relate to the marked suppression of IL-6 (by 75%) which is required for their maintenance through autocrine and paracrine pathways [97, 102]. Other factors that support GSC maintenance that are suppressed by HuR inhibition include TGF-β1 and 2, iNOS, and HIF1α [97, 103–106].
TAMs represent the third component of the GBM triad that relies on proper HuR function. TAMs, which are under the paracrine influence of GTCs and GSCs, are essential for creating a TME favorable for tumor progression [5, 107]. HuR positively regulates many of the factors secreted by TAMs that provide trophic support for GSCs and GTCs, modify the ECM for invasion and angiogenesis, and promote evasion of the immune system (Fig. 4 and Table 1). When HuR was selectively deleted in TAMs using CX3CR1-driven Cre recombinase, there was a significant attenuation of tumor growth and a prolongation of survival after intracranial injection of syngeneic GTCs [108]. Assessment of the immune microenvironment showed an overall reduction in TAMs but with an increase in the proportion of M1-like polarized cells (MHCIIhi, CD86+) and a decrease in M2-like polarization (MHCIIlo, CD206+). There was also an attenuation of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). PMN-MDSCs function as suppressors of T cell function and correlate with glioma grade and treatment resistance [109, 110]. In contrast, there was an overall increase infiltration of CD4+ T cells including Th1 and cytotoxic granzyme B+ and IFNγ+ effector cells which can directly kill cancer cells [111]. Interestingly, this cytotoxic activity is dependent on MHCII expression which is typically suppressed in GBM [112, 113]. However, in mice bearing microglia with ablated HuR, MHCII was increased not only in TAMs but also in GTCs [108]. The altered immune environment in these mice could relate to changes in molecular targets of HuR in TAMs including attenuation of CXCL1 and 2 (major chemokines for PMN-MDSCs), and an increase in CXCL10 which boosts recruitment of CD4+ cells and M1-like polarization [114]. Production of PD-L1, a transmembrane protein in TAMs (and GTCs) that suppresses anti-cancer T cell function, was also attenuated [107, 108, 115]. PD-L1 mRNA contains AREs in the 3 ‘UTR [116] and is markedly induced in TAMs by conditioned media from GTCs [108]. Both chemical inhibition and genetic deletion of HuR suppressed this induction [97, 108]. Other features of the TME that might be affected by HuR deletion in TAMs include the ECM (suppressed MMP2), angiogenesis (suppressed VEGF), and trophic support for GTCs and GSCs (suppressed VEGF, iNOS, IL-1β, IL-6, IL-8, and TGF-β1).
Taken together, HuR is an essential component of GBM progression through its regulation of intrinsic and secreted factors in the three major cellular components of GBM. The secreted factors govern a complex interplay in this triad and enable tumor cells to thrive in a hostile environment.
7. HuR inhibition as a therapeutic strategy in GBM
HuR as a potential therapeutic target was initially considered based on its upregulation in GBM and colon cancer, and its positive regulation of angiogenic factors [51, 86, 93]. Definitive proof of concept, however, was obtained from the discovery that shRNA-mediated HuR silencing in GTCs led to significant attenuation of tumor growth and invasion in vivo using an intracranial model [94]. The in vivo effect of HuR knockdown was observed by other laboratories [95]. Investigation of GTCs in culture show that HuR silencing reduces anchorage-dependent growth and increases apoptotic induction by chemotherapies used in the treatment of GBM including etoposide, topotecan and cisplatin [94]. Studies with xenograft-derived GTCs provide evidence that the extrinsic pathway of apoptosis is activated after HuR inhibition, with cleavage of caspase 8 and upregulation of death receptor 5 (DR5) [97]. A similar response was observed in HuR-silenced pancreatic cancer cells where HuR was thought to function as a stress-related translational repressor through binding to the 5’ UTR [117]. This role is distinct from ARE-mediated translational effects in the 3’ UTR and underscores the important concept that HuR function is not limited to AREs in the 3’ UTR. For example, HuR has been shown to bind certain internal ribosome entry site (IRES) in the 5’ UTR either to repress (e.g. insulin-like growth factor receptor and the anti-proliferative CDK inhibitor p27) or promote translation (e.g. the anti-apoptotic factor XIAP) [118–121].
In addition to the anti-cancer effects of HuR silencing in GTCs, ectopic overexpression of HuR enhances chemoresistance and expression of the anti-apoptotic factor Bcl-2 [94]. Taken together, these lines of evidence supporting an oncogenic role for HuR have fueled the search for small molecule inhibitors [87, 122]. Strategies have focused on two critical properties of HuR that are required for its regulatory function: RNA binding and cytoplasmic translocation (Fig. 5A). These two properties can be altered physiologically by posttranslational modifications to HuR, including phosphorylation, ubiquitination, methylation and proteolytic cleavage, at different amino acid residues within the RRMs and hinge region between RRMs 2 and 3 (reviewed in [42]). The first HuR inhibitor was discovered in a high-throughput screen using a fluorescence anisotropy-based assay to identify molecules that could disrupt HuR-ARE complexes [45]. The prototype inhibitors that emerged, including MS-444, disrupt HuR homodimerization, which appears to be a prerequisite for RNA-protein complex formation and ultimately translocation to the cytoplasm. While ARE binding can enhance multimerization, it does not appear to be an absolute requirement [38, 123]. In GBM, Filippova et al. observed extensive multimerization of cytoplasmic HuR in GBM clinical samples and postulated that this is an essential feature of HuR-driven oncogenesis [38]. They developed an elegant split-luciferase assay to detect and quantify HuR multimerization in GTCs (Fig. 5B). They fused the C- or N-terminal half of the firefly luciferase to HuR in two separate plasmids and transfected them into GTCs. Upon HuR dimerization, reconstitution of the luciferase protein produces luminescence with the addition of luciferin. With this assay they confirmed that MS-444 is a potent inhibitor of multimerization (Fig. 5B). Moreover, 15,16-dihydrotanshinone-I (DHTS), a compound identified by Provenzani et al. to inhibit complex formation between HuR and the TNF-α ARE [124], also disrupted multimerization (Fig. 5B) and suppressed GTC viability in cell culture [38]. Our preliminary studies indicate that DHTS suppresses glioma growth in mice (unpublished). These findings combined, with our other work [97], have implicated HuR multimerization as a critical step in glioma oncogenesis and a suitable target for developing therapies. Using this assay, Filippova et al. performed a high throughput screen of a synthetic library and identified a new family of small molecule inhibitors of HuR multimerization, with SRI-42127 being the prototype (Fig. 5B) [96]. Computational docking analysis predicts that SRI-42127 interacts with 4 key residues located in RRMs 1 and 2, suggesting that inhibition of multimerization results from a conformational change. Although the modeling predicts a disruption in RNA binding, this has not yet been tested. ARE binding and HuR multimerization appear to be closely linked, but it remains unclear which comes first.
Figure 5: Development of small molecules that block HuR multimerization.

(A) Based on studies with primary GBM tissues, there is extensive multimerization of cytoplasmic HuR which is an essential component to its oncogenic effect [38, 96]. (B) A split-luciferase assay was developed by Filippova and Nabors to detect HuR multimerization [38]. The N- and C-terminal halves of firefly luciferase were fused to HuR in separate plasmids which were then transfected into malignant glioma cells. Upon HuR dimerization, there is reconstitution of the luciferase protein as indicated by luminescence after addition of luciferin to the culture media. MS-444, a previously identified inhibitor of HuR dimerization [180] and lethal to malignant glioma cells [97], potently attenuates luminescence (shown to the right). A control plasmid expressing full-length luciferase is not affected. Likewise DHTS, which was shown by others to inhibit HuR-TNF-α ARE complex formation [124], also suppresses dimerization. The assay was then used in a high-throughput screen where a new class of dimerization inhibitors was discovered with SRI-42127 being the prototype [96].
The prototype inhibitor, SRI-42127, shows excellent penetration into the CNS (a barrier for new therapeutic molecules that often prevents advancement to the clinic) and suppresses GBM growth in vivo [96]. Interestingly, SRI-42127 potently suppresses HuR translocation in activated microglia and the production of key GBM-promoting factors associated with TAMs including IL-1β, IL-6, IL-8, iNOS, TNF-α, CCL2, CXCL1 and 2 [106]. Thus, the drug targets both GBM and the TME, adding potency to its anti-cancer effects. Because of the high amino acid sequence homology in the RRMs between HuR and neuronal members of ELAV [25], SRI-42127 may impact the function of neuronal ELAV members. DHTS for example, inhibits HuD-ARE complex formation [124]. It should be noted that other HuR inhibitors have been discovered that disrupt ARE complex formation or HuR trafficking but have not been tested in GBM and go beyond the scope of this review. The reader is directed to a recent review which nicely details the different candidate inhibitors, their putative mechanisms of action, and their anti-cancer effects [87].
8. Conclusions and perspectives
GBM remains a highly lethal tumor with little advancement in treatment options over the past three decades. Current strategies for treatment of glioblastoma rely on the modalities of surgery, radiation therapy, and primarily alkylating agent-based chemotherapy [125]. Although the Cancer Genome Atlas (TCGA) has identified potentially targetable oncogenes in GBM, such as EGFR, the extreme heterogeneity of the tumor substantially limits the clinical benefit of targeted therapies [126]. As covered in this review, HuR promotes multiple pro-oncogenic pathways within the major cellular components of GBM by increasing the stability and translational efficiency of mRNA transcripts encoding key molecular drivers of these pathways. Therapeutic targeting of HuR with a small molecule inhibitor such as SRI-42127 would therefore have a multipronged effect in suppressing GBM growth, invasion and resistance to therapy. This impact provides a strong rationale for advancing the discovery and refinement of small molecule HuR inhibitors for treatment of GBM and other cancers. The brain permeability of SRI-42127 overcomes a large obstacle in the treatment of GBM and holds promise for this class of inhibitors as adjuvant therapy to more conventional treatment modalities. Newer delivery methods using nanotechnology, convection-enhanced drug delivery, or intra-tumoral injections may further enhance this treatment approach [127].
Acknowledgements
This work is supported by NIH grants R01NS092651, R21NS111275–01, and Dept. of Veterans Affairs, BX001148 (PHK), R01 CA200624 (LBN), University of Alabama at Birmingham O’Neal Comprehensive Cancer Center Neuro-oncology Research Acceleration Fund (LBN), and NIA IRP, NIH (MG).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Nguyen HM, Guz-Montgomery K, Lowe DB, Saha D, Pathogenetic Features and Current Management of Glioblastoma, Cancers (Basel), 13 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Tamimi AF, Juweid M, Epidemiology and Outcome of Glioblastoma, in: De Vleeschouwer S (Ed.) Glioblastoma, Brisbane (AU), 2017. [PubMed] [Google Scholar]
- [3].Colwell N, Larion M, Giles AJ, Seldomridge AN, Sizdahkhani S, Gilbert MR, Park DM, Hypoxia in the glioblastoma microenvironment: shaping the phenotype of cancer stem-like cells, Neuro Oncol, 19 (2017) 887–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Lathia JD, Mack SC, Mulkearns-Hubert EE, Valentim CL, Rich JN, Cancer stem cells in glioblastoma, Genes Dev, 29 (2015) 1203–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Hambardzumyan D, Gutmann DH, Kettenmann H, The role of microglia and macrophages in glioma maintenance and progression, Nat. Neurosci, 19 (2016) 20–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Chen Z, Feng X, Herting CJ, Garcia VA, Nie K, Pong WW, Rasmussen R, Dwivedi B, Seby S, Wolf SA, Gutmann DH, Hambardzumyan D, Cellular and Molecular Identity of Tumor-Associated Macrophages in Glioblastoma, Cancer Res, 77 (2017) 2266–2278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Anderson P, Post-transcriptional control of cytokine production, Nat Immunol, 9 (2008) 353–359. [DOI] [PubMed] [Google Scholar]
- [8].Abdelmohsen K, Kuwano Y, Kim HH, Gorospe M, Posttranscriptional gene regulation by RNA-binding proteins during oxidative stress: implications for cellular senescence, Biol. Chem, 389 (2008) 243255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Barreau C, Paillard L, Osborne HB, AU-rich elements and associated factors: are there unifying principles?, Nuc Acids Res, 33 (2006) 7138–7150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Vasudevan S, Tong Y, Steitz JA, Switching from repression to activation: microRNAs can up-regulate translation, Science, 318 (2007) 1931–1934. [DOI] [PubMed] [Google Scholar]
- [11].Shaw G, Kamen R, A conserved AU sequence from the 3’ untranslated region of GM-CSF mRNA mediates selective mRNA degradation, Cell, 46 (1986) 659–667. [DOI] [PubMed] [Google Scholar]
- [12].Shen Z-J, Malter JS, Regulation of AU-Rich Element RNA Binding Proteins by Phosphorylation and the Prolyl Isomerase Pin1, Biomolecules, 5 (2015) 412–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Ihnatko R, Kubes M, Takacova M, Sedlakova O, Sedlak J, Pastorek J, Kopacek J, Pastorekova S, Extracellular acidosis elevates carbonic anhydrase IX in human glioblastoma cells via transcriptional modulation that does not depend on hypoxia, Int. J. Oncol, 29 (2006) 1025–1033. [PubMed] [Google Scholar]
- [14].Chen CY, Shyu AB, AU-rich elements: characterization and importance in mRNA degradation, Trends Biochem. Sci, 20 (1995) 465–470. [DOI] [PubMed] [Google Scholar]
- [15].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, Molecular Cell, 43 (2011) 340–352. [DOI] [PubMed] [Google Scholar]
- [16].Mukherjee N, Jacobs NC, Hafner M, Kennington EA, Nusbaum JD, Tuschl T, Blackshear PJ, Ohler U, Global target mRNA specification and regulation by the RNA-binding protein ZFP36, Genome Biol, 15 (2014) R12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].de Silanes IL, Zhan M, Lal A, Yang X, Gorospe M, Identification of a target RNA motif for RNA-binding protein HuR, PNAS, 101 (2004) 2987–2992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Bakheet T, Hitti E, Al-Saif M, Moghrabi WN, Khabar KSA, The AU-rich element landscape across human transcriptome reveals a large proportion in introns and regulation by ELAVL1/HuR, Biochim Biophys Acta, 1861 (2018) 167–177. [DOI] [PubMed] [Google Scholar]
- [19].Bakheet T, Williams BRG, Khabar KSA, ARED 3.0: the large and diverse AU-rich transcriptome, Nucl. Acids Res, 34 (2006) D111–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Wilusz CJ, Wilusz J, Bringing the role of mRNA decay in the control of gene expression into focus, Trends Genet, 20 (2004) 491–497. [DOI] [PubMed] [Google Scholar]
- [21].Gorospe M, Tominaga K, Wu X, Fahling M, Ivan M, Post-Transcriptional Control of the Hypoxic Response by RNA-Binding Proteins and MicroRNAs, Front. Mol. Neurosci, 4 (2011) 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Ma W-J, Cheng S, Campbell C, Wright A, Furneaux H, Cloning and Characterization of HuR, a Ubiquitously Expressed Elav-like Protein, J. Biol. Chem, 271 (1996) 8144–8151. [DOI] [PubMed] [Google Scholar]
- [23].Szabo A, Dalmau J, Manley G, Rosenfeld M, Wong E, Henson J, Posner JB, Furneaux HM, HuD a paraneoplastic encephalomyelitis antigen contains RNA binding domains and is homologous to elav and sex-lethal, Cell, 67 (1991) 325–333. [DOI] [PubMed] [Google Scholar]
- [24].Dalmau J, Furneaux HM, Gralla RJ, Kris MG, Posner JB, Detection of the anti-Hu antibody in the serum of patients with small cell lung cancer--a quantitative western blot analysis, Ann Neurol, 27 (1990) 544–552. [DOI] [PubMed] [Google Scholar]
- [25].Nabors LB, Furneaux HM, King PH, HuR, a novel target of anti-Hu antibodies, is expressed in non-neural tissues, Journal of Neuroimmunology, 92 (1998) 152–159. [DOI] [PubMed] [Google Scholar]
- [26].King PH, Levine TD, Fremeau RT Jr., Keene JD, Mammalian homologs of Drosophila ELAV localized to a neuronal subset can bind in vitro to the 3’ UTR of mRNA encoding the Id transcriptional repressor, J Neurosci, 14 (1994) 1943–1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].King PH, Redden D, Palmgren JS, Nabors LB, Lennon VA, Hu antigen specificities of ANNA-I autoantibodies in paraneoplastic neurological syndromes, J. Autoimmun, 13 (1999) 435–443. [DOI] [PubMed] [Google Scholar]
- [28].Levine TD, Gao F, King PH, Andrews LG, Keene JD, Hel-N1: an autoimmune RNA-binding protein with specificity for 3’ uridylate-rich untranslated regions of growth factor mRNAs, Mol. Cell. Biol, 13 (1993) 3494–3504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Yeap BB, Voon DC, Vivian JP, McCulloch RK, Thomson AM, Giles KM, Czyzyk-Krzeska MF, Furneaux H, Wilce MCJ, Wilce JA, Leedman PJ, Novel Binding of HuR and Poly(C)-binding Protein to a Conserved UC-rich Motif within the 3’-Untranslated Region of the Androgen Receptor Messenger RNA, J. Biol. Chem, 277 (2002) 27183–27192. [DOI] [PubMed] [Google Scholar]
- [30].Mukherjee N, Corcoran David L., Nusbaum Jeffrey D., Reid David W., Georgiev S, Hafner M, Ascano M Jr, Tuschl T, Ohler U, Keene Jack D., Integrative Regulatory Mapping Indicates that the RNA-Binding Protein HuR Couples Pre-mRNA Processing and mRNA Stability, Molecular Cell, 43 (2011) 327–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Ma WJ, Chung S, Furneaux H, The Elav-like proteins bind to AU-rich elements and to the poly(A) tail of mRNA, Nucleic Acids Res, 25 (1997) 3564–3569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Scheiba RM, de Opakua AI, Diaz-Quintana A, Cruz-Gallardo I, Martinez-Cruz LA, Martinez-Chantar ML, Blanco FJ, Diaz-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, 11 (2014) 1250–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Pabis M, Popowicz GM, Stehle R, Fernández-Ramos D, Asami S, Warner L, García-Mauriño SM, Schlundt A, Martínez-Chantar ML, Díaz-Moreno I, Sattler M, HuR biological function involves RRM3-mediated dimerization and RNA binding by all three RRMs, Nucleic Acids Res, 47 (2018) 1011–1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].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, 69 (2013) 373–380. [DOI] [PubMed] [Google Scholar]
- [35].Ripin N, Boudet J, Duszczyk MM, Hinniger A, Faller M, Krepl M, Gadi A, Schneider RJ, Šponer J, Meisner-Kober NC, Allain FH-T, Molecular basis for AU-rich element recognition and dimerization by the HuR C-terminal RRM, Proc. Natl. Acad. Sci, 116 (2019) 2935–2944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Toba G, White K, The third RNA recognition motif of Drosophila ELAV protein has a role in multimerization, Nucleic Acids Res, 36 (2008) 1390–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Diaz-Quintana A, Garcia-Maurino SM, Diaz-Moreno I, Dimerization model of the C-terminal RNA Recognition Motif of HuR, FEBS Lett, 589 (2015) 1059–1066. [DOI] [PubMed] [Google Scholar]
- [38].Filippova N, Yang X, Ananthan S, Sorochinsky A, Hackney JR, Gentry Z, Bae S, King P, Nabors LB, Hu antigen R (HuR) multimerization contributes to glioma disease progression, J. Biol. Chem, 292 (2017) 16999–17010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Balcerak A, Trebinska-Stryjewska A, Konopinski R, Wakula M, Grzybowska EA, RNA-protein interactions: disorder, moonlighting and junk contribute to eukaryotic complexity, Open Biol, 9 (2019) 190096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Fialcowitz-White EJ, Brewer BY, Ballin JD, Willis CD, Toth EA, Wilson GM, Specific Protein Domains Mediate Cooperative Assembly of HuR Oligomers on AU-rich mRNA-destabilizing Sequences, J. Biol. Chem, 282 (2007) 20948–20959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Fan XC, Steitz JA, HNS, a nuclear-cytoplasmic shuttling sequence in HuR, Proc. Natl. Acad. Sci. USA, 1998, pp. 15293–15298. [DOI] [PMC free article] [PubMed]
- [42].Grammatikakis I, Abdelmohsen K, Gorospe M, Posttranslational control of HuR function, Wiley Interdiscip Rev RNA, 8 (2017). [DOI] [PMC free article] [PubMed]
- [43].Dhamija S, Kuehne N, Winzen R, Doerrie A, Dittrich-Breiholz O, Thakur BK, Kracht M, Holtmann H, Interleukin-1 activates synthesis of interleukin-6 by interfering with a KH-type splicing regulatory protein (KSRP)-dependent translational silencing mechanism, J. Biol. Chem, 286 (2011) 33279–33288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Brooks SA, Blackshear PJ, Tristetraprolin (TTP): Interactions with mRNA and proteins, and current thoughts on mechanisms of action, Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, 1829 (2013) 666–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Meisner NC, Hintersteiner M, Mueller K, Bauer R, Seifert JM, Naegeli HU, Ottl J, Oberer L, Guenat C, Moss S, Harrer N, Woisetschlaeger M, Buehler C, Uhl V, Auer M, Identification and mechanistic characterization of low-molecular-weight inhibitors for HuR, Nat. Chem. Biol, 3 (2007) 508–515. [DOI] [PubMed] [Google Scholar]
- [46].Stoecklin G, Mayo T, Anderson P, ARE-mRNA degradation requires the 5’–3’ decay pathway, EMBO rep, 7 (2006) 72–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Fenger-Gron M, Fillman C, Norrild B, Lykke-Andersen J, Multiple processing body factors and the ARE binding protein TTP activate mRNA decapping, Mol Cell, 20 (2005) 905–915. [DOI] [PubMed] [Google Scholar]
- [48].Mitchell P, Petfalski E, Shevchenko A, Mann M, Tollervey D, The Exosome: A Conserved Eukaryotic RNA Processing Complex Containing Multiple 3’-->5’ Exoribonucleases, Cell, 91 (1997) 457–466. [DOI] [PubMed] [Google Scholar]
- [49].Zucconi BE, Wilson GM, Modulation of neoplastic gene regulatory pathways by the RNA-binding factor AUF1, Front. Biosci. (Landmark Ed), 16 (2011) 2307–2325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Bhattacharyya SN, Habermacher R, Martine U, Closs EI, Filipowicz W, Relief of microRNA-mediated translational repression in human cells subjected to stress, Cell, 125 (2006) 1111–1124. [DOI] [PubMed] [Google Scholar]
- [51].Nabors LB, Gillespie GY, Harkins L, King PH, HuR, an 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, 61 (2001) 2154–2161. [PubMed] [Google Scholar]
- [52].Demuth T, Reavie LB, Rennert JL, Nakada M, Nakada S, Hoelzinger DB, Beaudry CE, Henrichs AN, Anderson EM, Berens ME, MAP-ing glioma invasion: Mitogen-activated protein kinase kinase 3 and p38 drive glioma invasion and progression and predict patient survival, Mol Cancer Ther, 6 (2007) 1212–1222. [DOI] [PubMed] [Google Scholar]
- [53].Gurgis FM, Yeung YT, Tang MX, Heng B, Buckland M, Ammit AJ, Haapasalo J, Haapasalo H, Guillemin GJ, Grewal T, Munoz L, The p38-MK2-HuR pathway potentiates EGFRvIII-IL-1beta-driven IL-6 secretion in glioblastoma cells, Oncogene, 34 (2015) 2934–2942. [DOI] [PubMed] [Google Scholar]
- [54].Lopez de Silanes I, Lal A, Gorospe M, HuR: post-transcriptional paths to malignancy, RNA Biol, 2 (2005) 11–13. [DOI] [PubMed] [Google Scholar]
- [55].Tiedje C, Ronkina N, Tehrani M, Dhamija S, Laass K, Holtmann H, Kotlyarov A, Gaestel M, The p38/MK2-driven exchange between tristetraprolin and HuR regulates AU-rich element-dependent translation, PLoS Genet, 8 (2012) e1002977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].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, 23 (2003) 7177–7188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Lafarga V, Cuadrado A, Lopez de Silanes I, Bengoechea R, Fernandez-Capetillo O, Nebreda AR, p38 Mitogen-activated protein kinase- and HuR-dependent stabilization of p21(Cip1) mRNA mediates the G(1)/S checkpoint, Mol. Cell. Biol, 29 (2009) 4341–4351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Sandler H, Stoecklin G, Control of mRNA decay by phosphorylation of tristetraprolin, Biochem. Soc. Trans, 36 (2008) 491–496. [DOI] [PubMed] [Google Scholar]
- [59].Suswam E, Li Y, Zhang X, Gillespie GY, Li X, Shacka JJ, Lu L, Zheng L, King PH, Tristetraprolin down-regulates interleukin-8 and vascular endothelial growth factor in malignant glioma cells, Cancer Res, 68 (2008) 674–682. [DOI] [PubMed] [Google Scholar]
- [60].Stoecklin G, Stubbs T, Kedersha N, Wax S, Rigby WFC, Blackwell TK, Anderson P, MK2-induced tristetraprolin:14–3-3 complexes prevent stress granule association and ARE-mRNA decay, EMBO J, 23 (2004) 1313–1324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Suswam EA, Shacka JJ, Walker K, Lu L, Li X, Si Y, Zhang X, Zheng L, Nabors LB, Cao H, King PH, Mutant tristetraprolin: a potent inhibitor of malignant glioma cell growth, J. Neurooncol, 113 (2013) 195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Abdelmohsen K, Srikantan S, Kuwano Y, Gorospe M, miR-519 reduces cell proliferation by lowering RNA-binding protein HuR levels, Proc Natl Acad Sci U S A, 105 (2008) 20297–20302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Guha A, Ahuja D, Das Mandal S, Parasar B, Deyasi K, Roy D, Sharma V, Willard B, Ghosh A, Ray PS, Integrated Regulation of HuR by Translation Repression and Protein Degradation Determines Pulsatile Expression of p53 Under DNA Damage, iScience, 15 (2019) 342–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Xu F, Zhang X, Lei Y, Liu X, Liu Z, Tong T, Wang W, Loss of repression of HuR translation by miR-16 may be responsible for the elevation of HuR in human breast carcinoma, J. Cell. Biochem, 111 (2010) 727–734. [DOI] [PubMed] [Google Scholar]
- [65].Kojima K, Fujita Y, Nozawa Y, Deguchi T, Ito M, MiR-34a attenuates paclitaxel-resistance of hormone-refractory prostate cancer PC3 cells through direct and indirect mechanisms, Prostate, 70 (2010) 1501–1512. [DOI] [PubMed] [Google Scholar]
- [66].Guo X, Wu Y, Hartley RS, MicroRNA-125a represses cell growth by targeting HuR in breast cancer, RNA Biol, 6 (2009) 575–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Abdelmohsen K, Srikantan S, Yang X, Lal A, Kim HH, Kuwano Y, Galban S, Becker KG, Kamara D, de Cabo R, Gorospe M, Ubiquitin-mediated proteolysis of HuR by heat shock, EMBO J, 28 (2009) 1271–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Chu PC, Chuang HC, Kulp SK, Chen CS, The mRNA-stabilizing factor HuR protein is targeted by β-TrCP protein for degradation in response to glycolysis inhibition, J. Biol. Chem, 287 (2012) 43639–43650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Daks A, Petukhov A, Fedorova O, Shuvalov O, Kizenko A, Tananykina E, Vasileva E, Semenov O, Bottrill A, Barlev N, The RNA-binding protein HuR is a novel target of Pirh2 E3 ubiquitin ligase, Cell Death Dis, 12 (2021) 581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Lan Y, Xiao X, He Z, Luo Y, Wu C, Li L, Song X, Long noncoding RNA OCC-1 suppresses cell growth through destabilizing HuR protein in colorectal cancer, Nucleic Acids Res, 46 (2018) 5809–5821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Lucchesi C, Sheikh MS, Huang Y, Negative regulation of RNA-binding protein HuR by tumor-suppressor ECRG2, Oncogene, 35 (2016) 2565–2573. [DOI] [PubMed] [Google Scholar]
- [72].Mazroui R, Di Marco S, Clair E, von Roretz C, Tenenbaum SA, Keene JD, Saleh M, Gallouzi IE, Caspase-mediated cleavage of HuR in the cytoplasm contributes to pp32/PHAP-I regulation of apoptosis, J. Cell Biol, 180 (2008) 113–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Talwar S, Jin J, Carroll B, Liu A, Gillespie MB, Palanisamy V, Caspase-mediated cleavage of RNA-binding protein HuR regulates c-Myc protein expression after hypoxic stress, J. Biol. Chem, 286 (2011) 32333–32343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Srikantan S, Tominaga K, Gorospe M, Functional interplay between RNA-binding protein HuR and microRNAs, Current protein & peptide science, 13 (2012) 372–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Ahuja D, Goyal A, Ray PS, Interplay between RNA-binding protein HuR and microRNA-125b regulates p53 mRNA translation in response to genotoxic stress, RNA Biol, 13 (2016) 1152–1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Srikantan S, Gorospe M, HuR function in disease, Front. Biosci. (Landmark Ed), 17 (2012) 189–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Kim HH, Kuwano Y, Srikantan S, Lee EK, Martindale JL, Gorospe M, HuR recruits let-7/RISC to repress c-Myc expression, Genes Dev, 23 (2009) 1743–1748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Balkhi MY, Iwenofu OH, Bakkar N, Ladner KJ, Chandler DS, Houghton PJ, London CA, Kraybill W, Perrotti D, Croce CM, Keller C, Guttridge DC, miR-29 acts as a decoy in sarcomas to protect the tumor suppressor A20 mRNA from degradation by HuR, Science signaling, 6 (2013) ra63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Dormoy-Raclet V, Cammas A, Celona B, Lian XJ, van der Giessen K, Zivojnovic M, Brunelli S, Riuzzi F, Sorci G, Wilhelm BT, Di Marco S, Donato R, Bianchi ME, Gallouzi IE, HuR and miR-1192 regulate myogenesis by modulating the translation of HMGB1 mRNA, Nat. Commun, 4 (2013) 2388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Young LE, Moore AE, Sokol L, Meisner-Kober N, Dixon DA, The mRNA stability factor HuR inhibits microRNA-16 targeting of COX-2, Mol. Cancer Res, 10 (2012) 167–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Poria DK, Guha A, Nandi I, Ray PS, RNA-binding protein HuR sequesters microRNA-21 to prevent translation repression of proinflammatory tumor suppressor gene programmed cell death 4, Oncogene, 35 (2016) 1703–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Mukherjee K, Ghoshal B, Ghosh S, Chakrabarty Y, Shwetha S, Das S, Bhattacharyya SN, Reversible HuR-microRNA binding controls extracellular export of miR-122 and augments stress response, EMBO Rep, 17 (2016) 1184–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Shweiki D, Itin A, Soffer D, Keshet E, Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis, Nature, 359 (1992) 843–845. [DOI] [PubMed] [Google Scholar]
- [84].Desbaillets I, Diserens AC, Tribolet N, Hamou MF, Van Meir EG, Upregulation of interleukin 8 by oxygen-deprived cells in glioblastoma suggests a role in leukocyte activation, chemotaxis, and angiogenesis, J. Exp. Med, 186 (1997) 1201–1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Deininger MH, Weller M, Streffer J, Mittelbronn M, Meyermann R, Patterns of cyclooxygenase-1 and −2 expression in human gliomas in vivo, Acta Neuropathologica, 98 (1999) 240–244. [DOI] [PubMed] [Google Scholar]
- [86].Dixon DA, Tolley ND, King PH, Nabors LB, McIntyre TM, Zimmerman GA, Prescott SM, Altered expression of the mRNA stability factor HuR promotes cyclooxygenase-2 expression in colon cancer cells, J Clin Invest, 108 (2001) 1657–1665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Schultz CW, Preet R, Dhir T, Dixon DA, Brody JR, Understanding and targeting the disease-related RNA binding protein human antigen R (HuR), Wiley Interdiscip Rev RNA, 11 (2020) e1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Hanahan D, Robert A Weinberg, Hallmarks of Cancer: The Next Generation, Cell, 144 (2011) 646–674. [DOI] [PubMed] [Google Scholar]
- [89].Levy NS, Chung S, Furneaux H, Levy AP, Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR, J. Biol. Chem, 273 (1998) 6417–6423. [DOI] [PubMed] [Google Scholar]
- [90].Li X, Lu L, Bush DJ, Zhang X, Zheng L, Suswam EA, King PH, 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, 108 (2009) 1032–1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].King PH, RNA-binding analyses of HuC and HuD with the VEGF and c-myc 3’-untranslated regions using a novel ELISA-based assay, Nucleic Acids Res, 28 (2000) e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Zhu VF, Yang J, LeBrun DG, Li M, Understanding the role of cytokines in Glioblastoma Multiforme pathogenesis, Cancer Letters, 316 (2012) 139–150. [DOI] [PubMed] [Google Scholar]
- [93].Nabors LB, Suswam E, Huang Y, Yang X, Johnson MJ, King PH, Tumor necrosis factor alpha induces angiogenic factor up-regulation in malignant glioma cells: a role for RNA stabilization and HuR, Cancer Res, 63 (2003) 4181–4187. [PubMed] [Google Scholar]
- [94].Filippova N, Yang X, Wang Y, Gillespie GY, Langford C, King PH, Wheeler C, Nabors LB, The RNA-binding protein HuR promotes glioma growth and treatment resistance, Mol. Cancer Res, 9 (2011) 648–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Holmes B, Benavides-Serrato A, Freeman RS, Landon KA, Bashir T, Nishimura RN, Gera J, mTORC2/AKT/HSF1/HuR constitute a feed-forward loop regulating Rictor expression and tumor growth in glioblastoma, Oncogene, 37 (2018) 732–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Filippova N, Yang X, Ananthan S, Calano J, Pathak V, Bratton L, Vekariya RH, Zhang S, Ofori E, Hayward EN, Namkoong D, Crossman DK, Crowley MR, King PH, Mobley J, Nabors LB, Targeting the HuR Oncogenic Role with a New Class of Cytoplasmic Dimerization Inhibitors, Cancer Res, 81 (2021) 2220–2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Wang J, Hjelmeland AB, Nabors LB, King PH, Anti-cancer effects of the HuR inhibitor, MS-444, in malignant glioma cells, Cancer Biol Ther, 20 (2019) 979–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Filippova N, Yang X, King P, Nabors LB, Phosphoregulation of the RNA-binding protein Hu antigen R (HuR) by Cdk5 affects centrosome function, J. Biol. Chem, 287 (2012) 32277–32287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL, Yu JS, Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma, Mol Cancer, 5 (2006) 67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN, Glioma stem cells promote radioresistance by preferential activation of the DNA damage response, Nature, 444 (2006) 756–760. [DOI] [PubMed] [Google Scholar]
- [101].Ortensi B, Setti M, Osti D, Pelicci G, Cancer stem cell contribution to glioblastoma invasiveness, Stem Cell Res. Ther, 4 (2013) 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Wang H, Lathia JD, Wu Q, Wang J, Li Z, Heddleston JM, Eyler CE, Elderbroom J, Gallagher J, Schuschu J, MacSwords J, Cao Y, McLendon RE, Wang X-F, Hjelmeland AB, Rich JN, Targeting Interleukin 6 Signaling Suppresses Glioma Stem Cell Survival and Tumor Growth, Stem cells (Dayton, Ohio), 27 (2009) 2393–2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Tran AN, Boyd NH, Walker K, Hjelmeland AB, NOS Expression and NO Function in Glioma and Implications for Patient Therapies, Antioxid. Redox Signal, 26 (2017) 986–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Han J, Alvarez-Breckenridge CA, Wang Q-E, Yu J, TGF-β signaling and its targeting for glioma treatment, Am. J. Cancer Res, 5 (2015) 945–955. [PMC free article] [PubMed] [Google Scholar]
- [105].Heddleston JM, Hitomi M, Venere M, Flavahan WA, Yang K, Kim Y, Minhas S, Rich JN, Hjelmeland AB, Glioma stem cell maintenance: the role of the microenvironment, Curr. Pharm. Des, 17 (2011) 2386–2401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Chellappan R, Guha A, Si Y, Kwan T, Nabors LB, Filippova N, Yang X, Myneni AS, Meesala S, Harms AS, King PH, SRI-42127, a novel small molecule inhibitor of the RNA regulator HuR, potently attenuates glial activation in a model of lipopolysaccharide-induced neuroinflammation, Glia, n/a (2021). [DOI] [PMC free article] [PubMed]
- [107].Chen Z, Hambardzumyan D, Immune Microenvironment in Glioblastoma Subtypes, Front. Immunol, 9 (2018) 1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Wang J, Leavenworth JW, Hjelmeland AB, Smith R, Patel N, Borg B, Si Y, King PH, Deletion of the RNA regulator HuR in tumor-associated microglia and macrophages stimulates anti-tumor immunity and attenuates glioma growth, Glia, 67 (2019) 2424–2439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Gabrilovich DI, Myeloid-Derived Suppressor Cells, Cancer Immunol. Res, 5 (2017) 3–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Massara M, Persico P, Bonavita O, Mollica Poeta V, Locati M, Simonelli M, Bonecchi R, Neutrophils in Gliomas, Front. Immunol, 8 (2017) 1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Quezada SA, Simpson TR, Peggs KS, Merghoub T, Vider J, Fan X, Blasberg R, Yagita H, Muranski P, Antony PA, Restifo NP, Allison JP, Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts, J. Exp. Med, 207 (2010) 637–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Schartner JM, Hagar AR, Van Handel M, Zhang L, Nadkarni N, Badie B, Impaired capacity for upregulation of MHC class II in tumor-associated microglia, Glia, 51 (2005) 279–285. [DOI] [PubMed] [Google Scholar]
- [113].Qian J, Luo F, Yang J, Liu J, Liu R, Wang L, Wang C, Deng Y, Lu Z, Wang Y, Lu M, Wang JY, Chu Y, TLR2 Promotes Glioma Immune Evasion by Downregulating MHC Class II Molecules in Microglia, Cancer Immunol. Res, 6 (2018) 1220–1233. [DOI] [PubMed] [Google Scholar]
- [114].Karin N, Razon H, Chemokines beyond chemo-attraction: CXCL10 and its significant role in cancer and autoimmunity, Cytokine, 109 (2018) 24–28. [DOI] [PubMed] [Google Scholar]
- [115].Sun C, Mezzadra R, Schumacher TN, Regulation and Function of the PD-L1 Checkpoint, Immunity, 48 (2018) 434–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Coelho MA, de Carne Trecesson S, Rana S, Zecchin D, Moore C, Molina-Arcas M, East P, Spencer-Dene B, Nye E, Barnouin K, Snijders AP, Lai WS, Blackshear PJ, Downward J, Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA, Immunity, 47 (2017) 1083–1099 e1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Pineda DM, Rittenhouse DW, Valley CC, Cozzitorto JA, Burkhart RA, Leiby B, Winter JM, Weber MC, Londin ER, Rigoutsos I, Yeo CJ, Gorospe M, Witkiewicz AK, Sachs JN, Brody JR, HuR’s post-transcriptional regulation of Death Receptor 5 in pancreatic cancer cells, Cancer Biol Ther, 13 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Meng Z, Jackson NL, Choi H, King PH, Emanuel PD, Blume SW, Alterations in RNA-binding activities of IRES-regulatory proteins as a mechanism for physiological variability and pathological dysregulation of IGF-IR translational control in human breast tumor cells, J. Cell. Physiol, 217 (2008) 172–183. [DOI] [PubMed] [Google Scholar]
- [119].Meng Z, King PH, Nabors LB, Jackson NL, Chen CY, Emanuel PD, Blume SW, 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, 33 (2005) 2962–2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Kullmann M, Gopfert U, Siewe B, Hengst L, ELAV/Hu proteins inhibit p27 translation via an IRES element in the p27 5’UTR, Genes Dev, 16 (2002) 3087–3099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Durie D, Lewis SM, Liwak U, Kisilewicz M, Gorospe M, Holcik M, RNA-binding protein HuR mediates cytoprotection through stimulation of XIAP translation, Oncogene, 30 (2011) 1460–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Wang J, Guo Y, Chu H, Guan Y, Bi J, Wang B, Multiple Functions of the RNA-Binding Protein HuR in Cancer Progression, Treatment Responses and Prognosis, Int J Mol Sci, 14 (2013) 10015–10041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].David PS, Tanveer R, Port JD, FRET-detectable interactions between the ARE binding proteins, HuR and p37AUF1, RNA, 13 (2007) 1453–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].D’Agostino VG, Lal P, Mantelli B, Tiedje C, Zucal C, Thongon N, Gaestel M, Latorre E, Marinelli L, Seneci P, Amadio M, Provenzani A, Dihydrotanshinone-I interferes with the RNA-binding activity of HuR affecting its post-transcriptional function, Sci. Rep, 5 (2015) 16478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, R. European Organisation for, T. Treatment of Cancer Brain, G. Radiotherapy, G. National Cancer Institute of Canada Clinical Trials, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma, N Engl J Med, 352 (2005) 987–996. [DOI] [PubMed] [Google Scholar]
- [126].N. Cancer Genome Atlas Research, Comprehensive genomic characterization defines human glioblastoma genes and core pathways, Nature, 455 (2008) 1061–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Cha GD, Kang T, Baik S, Kim D, Choi SH, Hyeon T, Kim DH, Advances in drug delivery technology for the treatment of glioblastoma multiforme, Journal of controlled release : official journal of the Controlled Release Society, 328 (2020) 350–367. [DOI] [PubMed] [Google Scholar]
- [128].Agarwal P, Pajor MJ, Anson DM, Guda MR, Labak CM, Tsung AJ, Velpula KK, Elucidating immunometabolic targets in glioblastoma, American journal of cancer research, 7 (2017) 1990–1995. [PMC free article] [PubMed] [Google Scholar]
- [129].Tirapelli D, Lustosa IL, Menezes SB, Franco IM, Rodrigues AR, Peria FM, Marinho A, Serafini LN, Carlotti CG Jr., Tirapelli LF, High expression of XIAP and Bcl-2 may inhibit programmed cell death in glioblastomas, Arquivos de neuro-psiquiatria, 75 (2017) 875–880. [DOI] [PubMed] [Google Scholar]
- [130].Qiu B, Wang Y, Tao J, Wang Y, Expression and correlation of Bcl-2 with pathological grades in human glioma stem cells, Oncol. Rep, 28 (2012) 155–160. [DOI] [PubMed] [Google Scholar]
- [131].Trisciuoglio D, Tupone MG, Desideri M, Di Martile M, Gabellini C, Buglioni S, Pallocca M, Alessandrini G, D’Aguanno S, Del Bufalo D, BCL-XL overexpression promotes tumor progression-associated properties, Cell Death Dis, 8 (2017) 3216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Cho HR, Kumari N, Thi Vu H, Kim H, Park C-K, Choi SH, Increased Antiangiogenic Effect by Blocking CCL2-dependent Macrophages in a Rodent Glioblastoma Model: Correlation Study with Dynamic Susceptibility Contrast Perfusion MRI, Sci. Rep, 9 (2019) 11085–11085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Chang AL, Miska J, Wainwright DA, Dey M, Rivetta CV, Yu D, Kanojia D, Pituch KC, Qiao J, Pytel P, Han Y, Wu M, Zhang L, Horbinski CM, Ahmed AU, Lesniak MS, CCL2 Produced by the Glioma Microenvironment Is Essential for the Recruitment of Regulatory T Cells and Myeloid-Derived Suppressor Cells, Cancer Res, 76 (2016) 5671–5682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Graham JR, Hendershott MC, Terragni J, Cooper GM, mRNA Degradation Plays a Significant Role in the Program of Gene Expression Regulated by Phosphatidylinositol 3-Kinase Signaling, Mol. Cell. Biol, 30 (2010) 5295–5305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Kwan T, Floyd CL, Kim S, King PH, RNA Binding Protein Human Antigen R Is Translocated in Astrocytes following Spinal Cord Injury and Promotes the Inflammatory Response, J Neurotrauma, 34 (2017) 1249–1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Yu-Ju Wu C, Chen CH, Lin CY, Feng LY, Lin YC, Wei KC, Huang CY, Fang JY, Chen PY, CCL5 of glioma-associated microglia/macrophages regulates glioma migration and invasion via calcium-dependent matrix metalloproteinase 2, Neuro Oncol, 22 (2020) 253–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Pham K, Luo D, Liu C, Harrison JK, CCL5, CCR1 and CCR5 in murine glioblastoma: immune cell infiltration and survival rates are not dependent on individual expression of either CCR1 or CCR5, J Neuroimmunol, 246 (2012) 10–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Mao P, Bao G, Wang Y-C, Du C-W, Yu X, Guo X-Y, Li R-C, Wang M-D, PDZ-Binding Kinase-Dependent Transcriptional Regulation of CCNB2 Promotes Tumorigenesis and Radio-Resistance in Glioblastoma, Transl. Oncol, 13 (2020) 287–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Mahzouni P, Taheri F, An Immunohistochemical Study of Cyclin D1 Expression in Astrocytic Tumors and its Correlation with Tumor Grade, Iran J Pathol, 14 (2019) 252–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Liang H, Chen Z, Sun L, Inhibition of cyclin E1 overcomes temozolomide resistance in glioblastoma by Mcl-1 degradation, Mol. Carcinog, 58 (2019) 1502–1511. [DOI] [PubMed] [Google Scholar]
- [141].Liu L, Li T, Song G, He Q, Yin Y, Lu JY, Bi X, Wang K, Luo S, Chen Y-S, Yang Y, Sun B-F, Yang Y-G, Wu J, Zhu H, Shen X, Insight into novel RNA-binding activities via large-scale analysis of lncRNA-bound proteome and IDH1-bound transcriptome, Nucleic Acids Research, 47 (2019) 2244–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Tannous BA, Badr CE, A TNF-NF-κB-STAT3 loop triggers resistance of glioma-stem-like cells to Smac mimetics while sensitizing to EZH2 inhibitors, Cell death & disease, 10 (2019) 268–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Wang J, Wang H, Li Z, Wu Q, Lathia JD, McLendon RE, Hjelmeland AB, Rich JN, c-Myc is required for maintenance of glioma cancer stem cells, PLoS One, 3 (2008) e3769–e3769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Annibali D, Whitfield JR, Favuzzi E, Jauset T, Serrano E, Cuartas I, Redondo-Campos S, Folch G, Gonzalez-Junca A, Sodir NM, Masso-Valles D, Beaulieu ME, Swigart LB, Mc Gee MM, Somma MP, Nasi S, Seoane J, Evan GI, Soucek L, Myc inhibition is effective against glioma and reveals a role for Myc in proficient mitosis, Nat. Commun, 5 (2014) 4632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Qiu J, Shi Z, Jiang J, Cyclooxygenase-2 in glioblastoma multiforme, Drug Discov. Today, 22 (2017) 148–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Sengupta S, Jang BC, Wu MT, Paik JH, Furneaux H, Hla T, The RNA-binding protein HuR regulates the expression of cyclooxygenase-2, J. Biol. Chem, 278 (2003) 25227–25233. [DOI] [PubMed] [Google Scholar]
- [147].Young LE, Sanduja S, Bemis-standoli K, Pena EA, Price RL, Dixon DA, The mRNA Binding Proteins HuR and Tristetraprolin Regulate Cyclooxygenase 2 Expression During Colon Carcinogenesis, Gastroenterology, 136 (2009) 1669–1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Alafate W, Li X, Zuo J, Zhang H, Xiang J, Wu W, Xie W, Bai X, Wang M, Wang J, Elevation of CXCL1 indicates poor prognosis and radioresistance by inducing mesenchymal transition in glioblastoma, CNS Neurosci. Ther, 26 (2020) 475–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Matsye P, Zheng L, Si Y, Kim S, Luo W, Crossman DK, Bratcher PE, King PH, HuR promotes the molecular signature and phenotype of activated microglia: Implications for amyotrophic lateral sclerosis and other neurodegenerative diseases, Glia, 65 (2017) 945–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Liu C, Luo D, Reynolds BA, Meher G, Katritzky AR, Lu B, Gerard CJ, Bhadha CP, Harrison JK, Chemokine receptor CXCR3 promotes growth of glioma, Carcinogenesis, 32 (2011) 129–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Chen Z, Mou L, Pan Y, Feng C, Zhang J, Li J, CXCL8 Promotes Glioma Progression By Activating The JAK/STAT1/HIF-1α/Snail Signaling Axis, Onco Targets Ther, 12 (2019) 8125–8138. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- [152].Zhou J, Yi L, Ouyang Q, Xu L, Cui H, Xu M, Neurotensin signaling regulates stem-like traits of glioblastoma stem cells through activation of IL-8/CXCR1/STAT3 pathway, Cell. signalling, 26 (2014) 2896–2902. [DOI] [PubMed] [Google Scholar]
- [153].Suswam EA, Nabors LB, Huang Y, Yang X, King PH, IL-1β 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, 113 (2005) 911–919. [DOI] [PubMed] [Google Scholar]
- [154].Maru SV, Holloway KA, Flynn G, Lancashire CL, Loughlin AJ, Male DK, Romero IA, Chemokine production and chemokine receptor expression by human glioma cells: role of CXCL10 in tumour cell proliferation, J Neuroimmunol, 199 (2008) 35–45. [DOI] [PubMed] [Google Scholar]
- [155].Stevenson CB, Ehtesham M, McMillan KM, Valadez JG, Edgeworth ML, Price RR, Abel TW, Mapara KY, Thompson RC, CXCR4 expression is elevated in glioblastoma multiforme and correlates with an increase in intensity and extent of peritumoral T2-weighted magnetic resonance imaging signal abnormalities, Neurosurgery, 63 (2008) 560–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Al-Souhibani N, Al-Ghamdi M, Al-Ahmadi W, Khabar KSA, Posttranscriptional control of the chemokine receptor CXCR4 expression in cancer cells, Carcinogenesis, 35 (2014) 1983–1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Muralidharan R, Panneerselvam J, Chen A, Zhao YD, Munshi A, Ramesh R, HuR-targeted nanotherapy in combination with AMD3100 suppresses CXCR4 expression, cell growth, migration and invasion in lung cancer, Cancer Gene Ther, 22 (2015) 581–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Bellail AC, Tse MC, Song JH, Phuphanich S, Olson JJ, Sun SY, Hao C, DR5-mediated DISC controls caspase-8 cleavage and initiation of apoptosis in human glioblastomas, J. Cell. Mol. Med, 14 (2010) 1303–1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Xu H, Zong H, Ma C, Ming X, Shang M, Li K, He X, Du H, Cao L, Epidermal growth factor receptor in glioblastoma, Oncol. Lett, 14 (2017) 512–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Goldman CK, Kim J, Wong WL, King V, Brock T, Gillespie GY, Epidermal growth factor stimulates vascular endothelial growth factor production by human malignant glioma cells: a model of glioblastoma multiforme pathophysiology, Molecular biology of the cell, 4 (1993) 121–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Hong I-S, Stimulatory versus suppressive effects of GM-CSF on tumor progression in multiple cancer types, Exp. Mol. Med, 48 (2016) e242–e242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Curran CS, Evans MD, Bertics PJ, GM-CSF production by glioblastoma cells has a functional role in eosinophil survival, activation, and growth factor production for enhanced tumor cell proliferation, J Immunol, 187 (2011) 1254–1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Raineri I, Wegmueller D, Gross B, Certa U, Moroni C, Roles of AUF1 isoforms, HuR and BRF1 in ARE-dependent mRNA turnover studied by RNA interference, Nucleic Acids Res, 32 (2004) 1279–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Lo Dico A, Martelli C, Diceglie C, Lucignani G, Ottobrini L, Hypoxia-Inducible Factor-1α Activity as a Switch for Glioblastoma Responsiveness to Temozolomide, Frontiers in oncology, 8 (2018) 249–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Chen Y-L, Huang Y-L, Lin N-Y, Chen H-C, Chiu W-C, Chang C-J, Differential regulation of ARE-mediated TNFα and IL-1β mRNA stability by lipopolysaccharide in RAW264.7 cells, Biochem. Biophys. Res. Commun, 346 (2006) 160–168. [DOI] [PubMed] [Google Scholar]
- [166].Gadani SP, Cronk JC, Norris GT, Kipnis J, IL-4 in the brain: a cytokine to remember, J Immunol, 189 (2012) 4213–4219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Yarovinsky TO, Butler NS, Monick MM, Hunninghake GW, Early exposure to IL-4 stabilizes IL-4 mRNA in CD4+ T cells via RNA-binding protein HuR, J Immunol, 177 (2006) 4426–4435. [DOI] [PubMed] [Google Scholar]
- [168].Qi L, Yu H, Zhang Y, Zhao D, Lv P, Zhong Y, Xu Y, IL-10 secreted by M2 macrophage promoted tumorigenesis through interaction with JAK2 in glioma, Oncotarget, 7 (2016) 71673–71685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S, Significance of interleukin-13 receptor alpha 2–targeted glioblastoma therapy, Neuro-Oncology, 16 (2014) 1304–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Casolaro V, Fang X, Tancowny B, Fan J, Wu F, Srikantan S, Asaki SY, De Fanis U, Huang SK, Gorospe M, Atasoy UX, Stellato C, Posttranscriptional regulation of IL-13 in T cells: role of the RNA-binding protein HuR, J. Allergy Clin. Immunol, 121 (2008) 853–859 e854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].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, 33 (2005) 4813–4827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Wu D-M, Hong X-W, Wen X, Han X-R, Wang S, Wang Y-J, Shen M, Fan S-H, Zhuang J, Zhang Z-F, Shan Q, Li M-Q, Hu B, Sun C-H, Lu J, Zheng Y-L, MCL1 gene silencing promotes senescence and apoptosis of glioma cells via inhibition of the PI3K/Akt signaling pathway, IUBMB life, 71 (2019) 81–92. [DOI] [PubMed] [Google Scholar]
- [173].Akool E-S, Kleinert H, Hamada FMA, Abdelwahab MH, Forstermann U, Pfeilschifter J, Eberhardt W, Nitric Oxide Increases the Decay of Matrix Metalloproteinase 9 mRNA by Inhibiting the Expression of mRNA-Stabilizing Factor HuR, Mol. Cell. Biol, 23 (2003) 4901–4916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Liwak U, Jordan LE, Von-Holt SD, Singh P, Hanson JEL, Lorimer IA, Roncaroli F, Holcik M, Loss of PDCD4 contributes to enhanced chemoresistance in Glioblastoma multiforme through de-repression of Bcl-xL translation, Oncotarget, 4 (2013) 1365–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Gaur AB, Holbeck SL, Colburn NH, Israel MA, Downregulation of Pdcd4 by mir-21 facilitates glioblastoma proliferation in vivo, Neuro-oncology, 13 (2011) 580–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Wigington CP, Jung J, Rye EA, Belauret SL, Philpot AM, Feng Y, Santangelo PJ, Corbett AH, Post-transcriptional regulation of programmed cell death 4 (PDCD4) mRNA by the RNA-binding proteins human antigen R (HuR) and T-cell intracellular antigen 1 (TIA1), J. Biol. Chem, 290 (2015) 3468–3487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Litak J, Mazurek M, Grochowski C, Kamieniak P, Roliński J, PD-L1/PD-1 Axis in Glioblastoma Multiforme, International journal of molecular sciences, 20 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Kaminska B, Kocyk M, Kijewska M, TGF beta signaling and its role in glioma pathogenesis, Adv. Exp. Med. Biol, 986 (2013) 171–187. [DOI] [PubMed] [Google Scholar]
- [179].Treps L, Perret R, Edmond S, Ricard D, Gavard J, Glioblastoma stem-like cells secrete the pro-angiogenic VEGF-A factor in extracellular vesicles, Journal of extracellular vesicles, 6 (2017) 1359479–1359479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Meisner N-C, Filipowicz W, Properties of the Regulatory RNA-Binding Protein HuR and its Role in Controlling miRNA Repression in: Großhans H (Ed.), Springer; New York: 2011, pp. 106–123. [DOI] [PubMed] [Google Scholar]
