Abstract
Cytokines and growth factors regulate cell proliferation, differentiation, migration and apoptosis, and play important roles in coordinating growth signal responses during development. The expression of cytokine genes and the signals transmitted through cytokine receptors are tightly regulated at several levels, including transcriptional and post-transcriptional levels. A majority of cytokine mRNAs, including growth factor transcripts, contain AU-rich elements (AREs) in their 3’ untranslated regions that control gene expression by regulating mRNA degradation and changing translational rates. In addition, numerous proteins involved in transmitting signals downstream of cytokine receptors are regulated at the level of mRNA degradation by GU-rich elements (GREs) found in their 3’ untranslated regions. Abnormal stabilization and overexpression of ARE or GRE-containing transcripts had been observed in many malignancies, which is a consequence of the malfunction of RNA-binding proteins. In this review, we briefly summarize the role of AREs and GREs in regulating mRNA turnover to coordinate cytokine and growth factor expression, and we describe how dysregulation of mRNA degradation mechanisms contributes to the development and progression of cancer.
Keywords: cytokine and growth factor signaling, post-transcriptional gene regulation, ARE, GRE, mRNA stability, cancer
1. Introduction
In addition to regulating cell proliferation, differentiation, and apoptosis, cytokines and growth factors also play important roles in coordinating growth signal responses during development. The ability of cytokines to affect cellular self-renewal capacity, migration, senescence, or apoptosis is often impaired in cancer. PubMed citations include approximately 7000 publications related to abnormal cytokine and growth factor signaling in cancer. Researchers have shown altered levels of expression or function of numerous cytokines, including growth factors and chemokines in malignant tissues relative to healthy tissues (reviewed in [1, 2]).
The expression of cytokines and growth factor signaling proteins are highly regulated at transcriptional and post-transcriptional levels [3, 4]. Cytokine and growth factor receptors and components of downstream signaling pathways are frequently overexpressed in cancer cells through abnormal mRNA stabilization, which promotes uncontrolled protein translation [5, 6]. Thus, it is crucial that the expression of cytokines and downstream signaling pathways are managed through multiple molecular mechanisms, including tightly regulated control of mRNA half-life [7, 8].
2. Levels of Post-transcriptional regulation
Expression of mammalian mRNA is regulated at multiple post-transcriptional levels that include splicing, cap addition, polyadenylation, transport, localization, degradation, and translation. mRNA molecules move from nucleus to the cytoplasm within messenger ribonucleoprotein (mRNP) complexes, dynamically associating with RNA-binding proteins (RNA-BPs) that bind to conserved cis-elements found in subsets of coordinately regulated transcripts (reviewed in [9, 10]). In the cytoplasm, the association of specific RNA-BPs with subsets of mRNAs containing conserved regulatory cis-elements coordinates the fate of these bound transcripts through post-transcriptional processes such as translation [11, 12], storage in stress granules [13], or mRNA decay (reviewed in [14, 15]). In addition to RNA-BPs, mRNA fate is also controlled by microRNAs (miRNAs) and non-coding RNAs that bind to cis-elements in mRNA (reviewed in [16–20]). Expression of cytokines and growth factor signaling molecules are most prominently regulated at post-transcriptional levels by conserved sequences found in the 3’ untranslated regions (3’UTRs) of their transcripts.
3. Regulation of cytokine and growth factor signaling by AREs
AREs are conserved sequences found in the 3’UTR of certain transcripts, including numerous cytokine, chemokine, and growth factor transcripts. AREs regulate post-transcriptional processes such as mRNA degradation and translation by binding to ARE-BPs that recruit other proteins to the mRNA that mediate post-transcriptional events. The sequence characteristics and decay patterns of different AREs allowed them to be initially categorized into three classes, based on the degradation kinetics and the number of overlapping AUUUA pentamers within the 3’UTR of a transcript [21]. An intensive genome-wide bioinformatics approach was used to classify ARE-containing transcripts into 5 clusters based on the number of overlapping AUUUA pentamers [22–24]. Cluster I AREs, which contain 5 overlapping AUUUA repeats, are enriched in secreted proteins, such as cytokines, chemokines, and growth factors, and are involved in the growth of hematopoietic and immune cells. The other ARE clusters are found within a diverse set of signaling transcripts. Remarkably, ARE-containing transcripts compose less than 8% of the human transcriptome [25], but they are highly enriched in transcripts involved in cytokine networks, found in up to 80% of transcripts within the cytokine and growth factor groups. AREs function to mediate mRNA degradation through their interaction with ARE-BPs, which compete with each other for ARE binding. Certain ARE-BPs such as ZFP36 (also called TTP), KSRP, or HNRNPD (AUF1) bind to AREs and mediate transcript degradation [26–28], while other ARE-BPs, such as ELAVL1 (also called HuR) or GAPDH [29], bind to AREs and mediate transcript stabilization, possibly by preventing binding by ARE-BPs that promote decay [30].
AREs play important roles in turning off the expression of immune activation genes during the resolution phase of immune responses (reviewed in [7, 31, 32]). For example, T cell receptor stimulation of human T lymphocytes induces expression of ARE-containing cytokine transcripts, such as IFNγ, IL2, IL4, TNFα, CSF etc., through increased transcription. This transcriptional induction is followed by transcript degradation mediated by the AREs found in the 3’UTRs of these transcripts [33, 34]. Premature post-transcriptional silencing of cytokine expressions through AREs may lead to an anergic, self-reactive T cell phenotype, which was observed in inflammatory types of cancer [35].
Cell activation can induce the expression of RNA-BPs (e.g. ZFP36, ZFP36L2, HNRNPD) that later bind to ARE-containing cytokine transcripts and recruit the cellular mRNA degradation machinery (e.g. deadenylases, exoribonucleases, or decapping enzymes) to the transcripts [36–38]. A subset of ARE-containing cytokine transcripts, such as IL6 and CXCL8, form stem-loop structures with double-stranded regions that activate exonucleases or endonucleases (e.g. RNase L) that degrade the transcripts [39–42]. In malignant cells, a number of ARE-containing cytokine the mRNAs are constitutively stable, suggesting that the function of the mRNA decay machinery in cancer is altered [43, 44]. The function of AREs and ARE-BPs can be altered due to atypical expression of ARE-BPs, post-translational alterations, mutations in binding regions, or dysregulated interplay with miRNAs, leading to changes in ARE-BP binding affinities [45–47].
4. ARE-BPs in cancer
The destabilizing functions of AREs and their ability to bind to distinct ARE-BPs differ in diseased versus healthy cells. The ELAVL1 and ZFP36 ARE-BPs have been shown to compete with one another for certain ARE-containing transcripts and exert opposite effects on the stability of ARE-containing mRNAs [48]. According to recent observation, ELAVL1 and ZFP36 share more than fifty percent of 3′UTRs target sites [49]. Binding by ELAVL1 promotes mRNA stabilization and upregulation of ARE-containing genes [50, 51], whereas, binding by ZFP36 promotes mRNA degradation and down-regulates the expression of ARE-containing genes [49, 52]. Remarkably, the expression and function of ELAVL1 is increased, but the function of ZFP36 is almost completely abrogated, in numerous types of tumors (reviewed in [53, 54]). Decreased function of ZFP36, ZFP36L1, and ZFP36L2, combined with elevated function of ELAVL1 result in increased expression of cytokine genes that promote cell growth and angiogenesis [55–58]. In humans, single nucleotide polymorphisms in ZFP36 and ELAVL1 genes are associated with poor outcomes in cancer patients [59, 60]. For the sake of brevity of this review, we shall focus on two ARE-BPs: ELAVL1 and ZFP36, as the malfunction of either, or both, has been reported as a trait of acquisition of tumorigenic and pro-metastatic properties (Fig. 1).
Fig. 1.
Alterations in post-transcriptional regulatory networks during tumorigenesis. Signals transmitted through prominent receptors that promote tumorigengesis, such as TGFβ growth factor receptors (which are also protein tyrosine kinases), WNT/β-catenin, and NOTCH, result in alterations in RNA-binding protein expression and function. These lead to dysregulation of mRNA stability/translation of major ARE- and GRE-containing transcripts that encode markers of EMT (described in text). Changes in expressions of these ARE- and GRE-containing transcripts cooperatively contribute to the overall phenotypic outcomes of cellular networks, such as feed-back loop activation of growth factor signaling pathways, cell proliferation, disruption of adherence junction, migration, invasion, and metastases.
4.1 ELAVL1 promotes tumor growth
Overexpression of the stabilizing protein ELAVL1 is strongly associated with proliferation: in cancer cell models, mice and in humans [61–66]. In fact, the link between ELAVL1 and cancer was initially discovered through observations of increased TNFα, VEGF, TGFβ, COX2, and CXCL8 mRNA expression in colon or brain cancer cells [67–69]. ELAVL1 gets rapidly phosphorylated following exposure to different stress stimuli, which may be an important mechanism utilized by cancer cells to keep up with cytotoxic stresses [70, 71]. For example, in colon carcinoma cells, the phosphorylation of ELAVL1 by PKC isozymes promotes COX2 overexpression through increased mRNA stability [72]. In hypoxic conditions, cytoplasmic localization and ARE-binding activity of ELAVL1 increases, leading to expression of pro-angiogenic mRNAs, likely due to activation of ELAVL1 by PKC and other kinases [73–77]. Moreover, ELAVL1 promotes chemo-resistance by binding to and stabilizing HIF1a and other hypoxia-associated mRNAs [78–80]. Interestingly, the mTOR inhibitor, rapamycin, inhibits ELAVL1 phosphorylation by PKC and decreases VEGF mRNA stability, suggesting that rapamycin might inhibit production of VEGF through its effect on ELAVL1, and thereby prevents neoplastic angiogenesis [81].
ELAVL1 increases not only pro-angiogenic, but also pro-inflammatory cytokine expression [82]. ELAVL1 is known to stabilize a number of pro-inflammatory mRNAs, and the overexpression of ELAVL1 in inflammatory types of glioblastoma, breast or lung cancer, is a risk factor for rapid disease progression [83–86]. Paradoxically, overexpression of ELAVL1 in breast cancer xenografts in athymic mice led to significant retardation of tumor growth, perhaps due to lack of angiogenesis in this xenograft model [87].
Overall, most existing data suggests that increased expression or cytoplasmic localization of ELAVL1 is associated with uncontrolled cell growth and chemoresistance [88–90]. In addition, the progression of cancer requires pro-inflammatory and pro-angiogenic signals from non-cancerous cell types within the tumor microenvironment, and these cancer-promoting signals may also be influenced by ELAVL1 [91]. Furthermore, genetic deletion of ELAVL1 in thymocytes [92] and myelocytes [93] predisposed mice to exaggerated inflammatory responses and inflammation-driven oncogenesis, likely due to malfunction in the cancer immune surveillance mechanism, which allows the formation of pro-tumorigenic milieu.
4.2 ZFP36 is a tumor suppressor
In contrast to ELAVL1, whose expression is upregulated in numerous forms of cancer, down-regulation of ZFP36 and ZFP36 family member’s expression or function was documented in many types of cancers [94, 95]. In fact, low ZFP36/ELAVL1 mRNA ratios correlate with high levels of the mitotic ARE-mRNA signature in a number of solid cancers [47]. ZFP36 functions to prevent the pathological overexpression of pro-inflammatory mRNAs that are associated with tumorigenesis, including IFNγ, IL1, IL2, IL6, IL8, IL10, IL16, IL17, IL23, TNFα, TGFβ, VEGF, and CSF1 [96–103]. These cytokine mRNAs are normally produced by immune and non-immune cells for a very short period of time in response to inflammatory signals, but are constitutively overexpressed in various types of tumors. The excessive production of cytokines in cancer has been attributed to absence or malfunction of ZFP36 (through transcriptional or post-translational mechanisms), which renders malignant cells unable to degrade ARE-containing transcripts in timely manner [104–107]. Thus, ZFP36 deficiency in tumors and immune cells may account for overproduction of these cytokines in the tumor microenvironment [108–110], leading to tumor cellular proliferation, vascularization and metastasis (Fig. 1). In support of this statement, knockdown of ZFP36 from tumors resulted in increased secretion of pro-inflammatory cytokines and accelerated tumor growth [104, 111]. In contrast, exogenous overexpression of functional ZFP36 triggered apoptosis of invasive breast cancer cell lines [112] or induced retardation of tumor growth and decreased metastatic potential in different cancer cell models (e.g. breast, cervical or colon carcinoma cell lines) [113]. The cancer therapeutic agent, Sorafenib, causes re-expression of ZFP36 in melanoma cells, which led to decreased expression of pro-angiogenic ARE-containing cytokines [114]. Overall, current literature supports the notion that the function of ZFP36 appears to counteract the cancer promoting properties of ELAVL1. In cancer cells, deficiency of ZFP36 is often seen in combination with increased expression/function of ELAVL1 [115–117]. Unbalanced expression of these two ARE-BPs appears to drive uncontrolled growth and contributes to tumorigenesis. Future research and development toward pharmacological agents that inhibit ELAVL1 and increase expression of ZFP36 may allow favorable control of post-transcriptional ARE networks and could potentially be used to treat devastating types of cancer, including gliomas and invasive types of breast, ovarian, or pancreatic cancers [118–122].
5. Regulation of cytokine and growth factor networks by GREs
Studies regarding AREs and ARE-BPs have led to the identification of other cis-elements and proteins that globally regulate mRNA decay [123]. For example, bioinformatic analyses of short-lived transcripts expressed in different cell types led our research team to identify the GRE sequences as highly enriched in the 3’UTRs of mRNAs that encode growth-promoting proteins [124, 125]. The GRE motif binds to the protein CELF1, leading to the decay of GRE-containing mRNA. These GRE-containing transcripts have variable nucleotide repeats, that can be defined as UGU[G/U]UGU[G/U]UGU [126]. More recent computational studies identified a number of mature miRNAs containing a 5’-UGUGU-3’ motif, which is known to stimulate of innate immune responses [127]. These miRNAs could also directly bind to CELF1 and affect mRNA stability and cellular protein expression [128]. Of note, GRE-containing mRNAs encode numerous transcripts involved in cytokine and growth signaling pathways that are binding targets of CELF1 [7, 129]. In primary cells, CELF1 binds and regulates expression of transcripts encoding hundreds of activators of cellular proliferation, presumably maintaining cell transitions through appropriate developmental stages [130–132]. We identified that in response to immune stimuli, CELF1 becomes phosphorylated, which prevents CELF1 binding to mRNA. This leads to transient stabilization and upregulation of GRE-containing transcripts, including components of cytokine and growth factor signaling [7, 130] [133]. Thus, CELF1 functions in normal cells to down-regulate the expression of GRE-containing transcripts that promote cell growth, including multiple components of cytokine and growth factor signaling pathways [134].
6. CELF1 expression and function in cancer
There are inconsistent results about levels of CELF1 protein expression in different types of malignant tissues, varying from overexpression to no changes [135–139]. A bioinformatics search using Oncomine did not reveal significant difference in CELF1 mRNA expression between normal and various human cancer transcriptomes (oncomine.org). CELF1 function, however, may be changed even if the protein levels are not altered in malignant cells, contributing to dysregulation of cytokine signaling. Our data indicate that in HeLa cells and malignant leukemia and lymphoma cell lines, CELF1 is unable to bind to mRNA targets identified in normal T cells, even though the target mRNAs were expressed in each cell type and the CELF1 protein levels did not differ. This failure to bind to target transcripts by CELF1 in malignant cell lines, might be explained by post-translational alterations of this protein (specifically, phosphorylation), and also increased retention to the nucleus, which correlates with stabilization and increased expression of GRE-containing target transcripts in the cytoplasm [126, 137]. These stabilized transcripts included transcription factors, cytokines and growth factor signaling molecules such as STAT5, JUN, CEBPBα/β, IL15, IL1RB, IFNγ, beta-catenin, and CCR5,7 [137, 140, 141]. We found that CELF1 was expressed in a hyper-phosphorylated form in these cancer cell lines, and this phosphorylation prevented it from binding to and degrading GRE-containing transcripts that control cell growth. Unexpectedly, we also identified that hyper-phosphorylation of CELF1 caused it to gain the ability to bind to and facilitate the degradation of another subset of GRE-containing transcripts that encode suppressors of proliferation (e.g. SOCS5, TNFRSF4, PIAS1, NKIRAS1). It is unknown how exactly CELF1 switches preferences for binding to mRNA targets, but this switch seems to be related to the phosphorylation state of CELF1. Thus, additional studies are needed to determine if post-translational modification of CELF1 causes dysregulation of the GRE network in cancer cells.
In addition to its role in mRNA degradation, CELF1 regulates many other aspects of mRNA metabolism. In fact, it was first discovered in patients suffering from myotonic dystrophy type 1 (DM1), a toxic RNA disease [142]. Links between DM1 and aberrant CELF1 function prompted clinical investigations of cancer incidence in individuals suffering from this inherited disorder. These studies revealed an increased overall risk of cancer in DM1 patients, and specifically thyroid cancer and choroidal melanoma [143–145]. In the general population, the association between the abnormal function of CELF1 and myelogenous leukemias [146], as well as hepatic [147], and mammary [148] cancers have been described, with cancer pathogenesis linked to dysregulation of C/EBPβ and p21 (CDKN1A) mRNA expression by CELF1. Thus, CELF1 dysregulation is a common feature of neoplastic growth and proliferation.
7. Cross-talk between ARE and GRE networks in cancer
The network of ARE-containing transcripts encodes various cytokines and growth factors that depend on circuits of downstream GRE-containing signaling transcripts to function (Fig. 2A,B). Furthermore, many GRE-containing CELF1 target transcripts are also targets of the ARE-BP ELAVL1, indicating that CELF1 and ELAVL1 may have opposing effects on target RNA stability or translation [149]. RNA recognition motifs for CELF1 require (GU)n or GUUUU repeats, whereas the recognition sequence for ELAVL1 is more broad, and ELAVL1 binds to a variety of AU/GU-rich sequences, including a poly-U sequence [150, 151]. Thus, CELF1 and ELAVL1 often compete for G/U-rich binding sites on a subset of target transcripts. We compared GRE-containing transcripts that were CELF1 targets in normal T cells [130], but lost binding to CELF1 in malignant cell lines [126, 137, 141], with ELAVL1 targets identified by in similar malignant cell types [152]. We found numerous transcripts that appeared to switch their binding preferences from CELF1 to ELAVL1 (see Fig. 2A,B). Depending on environmental conditions or cell type, these two proteins can change their preferred localization from the nucleus to the cytoplasm or vice versa, which affects their availability to bind to and regulate target mRNAs [153–156]. Thus, depending on which protein is more abundant in the cytoplasm, and on which protein has a higher affinity for the GU-or-U-rich binding site in a given target transcript, transcripts may undergo stabilization and translation versus decay. The increased abundance of phosphorylated cytoplasmic ELAVL1 in cancer cells may promote stabilization of numerous GU-or-U-rich mRNAs, especially in the setting of CELF1 inactivity due to hyper-phosphorylation [137].
Fig. 2.
AREs and GREs in growth factor signaling.
A. Schematic representation of tumor growth factor receptors (TGFR), vascular endothelial, epidermal, platelet-derived, or insulin growth factor receptors (VEGFR, EGFR, IGFR, PTDGFR) signaling. Signals transmitted through the tyrosine kinase family (RTK) receptors are directed through several major kinase signaling cascades, such as RAS/RAF/MEK/ERK and PI3/Akt/mTOR/GSK3 pro-survival pathways.
B. Schematic representation of NOTCH and WNT/β-catenin signal-transduction components that differentially function in a growth factor- and hypoxia- dependent manner. Frizzle and NOTCH receptors transduce a range of intracellular signals that alter the rate of proliferation and production of growth factors and markers of EMT through the post-transcriptional mechanisms. GRE-containing transcripts (shown in grey) represent direct binding targets of CELF1 in normal cells, as determined by RNA-immunoprecipitation assays [130, 131]. These transcripts exhibit a switch in their binding preferences from CELF1 in normal cells [130, 131] to ELAVL1 in malignant cells [152]. Transcripts in yellow contain AREs. Nodes labeled as ZFP36, ELAVL1, CELF1, are experimentally validated functional mRNA targets of ZFP36, ELAVL1, or CELF1. Pentagon-shaped nodes represent transcripts encoding kinases. Arrows indicate direct interactions and/or activations. This network diagram was built using Ingenuity Pathway Assistant Software.
We proposed a model as such: In normal cells, many cytokine and growth factor signaling transcripts are bound to CELF1 and targeted for degradation after they are transiently expressed following cellular activation; In cancer cells, CELF1 becomes hyper-phosphorylated and is rendered unable to bind to certain GRE-containing transcripts [157]. The increased levels of cytoplasmic ELAVL1 and enhanced binding affinity of ELAVL1 for mRNA, in cancer cells, leads to increased binding by ELAVL1 to GRE-containing mRNAs, stabilizing them and allowing them to be translated [158, 159]. Although further evidence is needed, this model could explain the overexpression of certain ARE and GRE-containing transcripts in malignancy.
8. Post-transcriptional regulation of growth factor signaling in cancer
Tumor cells have been shown to acquire the ability to produce growth-promoting cytokines and to overexpress growth factor receptors [160]. Growth factor – induced cell proliferation, adhesion and migration often depend on ARE and GRE post-transcriptional regulatory networks. The TGFβ signaling cascade, for example, is encoded by ARE- and GRE-containing mRNAs (Fig. 2A). Activated by TGFβ, TGF receptors (TGFR) and downstream SMAD proteins regulate the transcription of a number of GRE-containing mRNAs (ex. CITED2, SNAILs, Cadherins, occludin, TWIST, CDKN1A, ETS1/2, TGFBR1, myc, jun, fos, etc.), which in turn become a subject of post-transcriptional regulation by CELF1 or ELAVL1 once they leave the nucleus [161–163]. TGFβ treatment causes nuclear ELAVL1 to translocate into the cytoplasm, where it binds to TGFβ mRNA and increases its stability, completing a positive feedback loop [164, 165]. In pathological conditions, GFRs also signal through a RAS oncogenic pathway, which leads to upregulation of a large set of ARE-containing transcripts and specifically VEGF mRNA, due to dysfunctional mRNA degradation and translational upregulation [166]. Overproduced VEGF acts on vascular endothelial cells to increase vascular permeability via tyrosine kinase receptors (TKR), which heightens the metastatic potential [167]. In addition to VEGF mRNA, ARE and GRE-containing transcripts encoding components of cell adhesion and motility pathways (e.g. ICAM1, VCAM1, E-selectin, β-actin or catenin) and proteins with enzymatic activities (e.g. heparanases, matrix metallo-proteinases MMP2,7,8,9 or uPA (PLAU), PIA) are stabilized and upregulated in cancer [118, 168–171] (see fig. 2B). The RAS-mTOR pathway is a well-established signaling axis that modulates the proliferation and survival of many cell types downstream of growth factor receptors. Abnormal growth factor signaling through RAS/RAF/MEK/ERK and PI3/Akt/mTOR pro-survival pathways can fuel cancer growth and metastases, in part through posttranscriptional mechanisms [157, 172, 173]. Since ARE and GRE-containing transcripts encode important regulators of apoptosis and the cell cycle that function downstream of growth factor receptors, transformed cells may usurp these post-transcriptional networks to their advantage as they become malignant [174–177] (Fig. 2A).
Key regulators of epithelial-to-mesenchymal transitions (EMT), such as SNAIL, β-catenin, and TWIST are encoded by ARE and GRE-containing molecules, and are regulated through post-transcriptional effect of ELAVL1, ZFP36, and CELF1 [178–181]. EMTs are driven by many soluble growth factors, including FGFs, HGF, EGF, PDGFC, IGF, and their cognate receptors [135, 182]. For example, activation of the EGFR pathway in triple-negative breast cancer increases the cytoplasmic localization of ELAVL1, leading to stabilization and translation of ARE-containing mRNAs such as CA9, SNAI2, β-catenin and IL6, contributing to the development of a stem cell-like and aggressive basal-like breast cancer phenotype [183]. Tumor cells often overexpress growth factor mRNAs through ELAVL1-directed stabilization, thereby creating positive auto-feedback loops in which more growth-promoting cytokines are produced and become available in the tumor microenvironment. On the background of poor anti-tumor immune response [184, 185], hypoxia [186], and proliferative signals from extracellular matrix components (e.g. Wnt and JAG/DLL), these feedback loops foster the disruption of adherence junctions and promote tumor angiogenesis and metastases (Fig. 1) [187–190]. Under normal conditions, ZFP36 and ZFP36L1 [191, 192] destabilize and degrade ARE-containing growth factor signaling transcripts, but in malignant cells these transcripts are stable and upregulated [43]. Given that cancer cells are often deficient in ZFP36, mRNA stabilization mechanisms could drive overexpression of growth-promoting networks in malignant tissues [193, 194]. Figure 2B depicts transcripts encoding NOTCH and WNT/β-catenin signaling components that differentially function in a growth factor- and hypoxia- dependent manner [195]. As can be seen, GRE-containing transcripts (grey transcripts) encode components of these signaling pathways downstream from Frizzle and NOTCH receptors. Overall, these receptors transduce a range of intracellular signals that alter the rate of proliferation and production of other cytokines, growth factors (e.g. IL1,6,8, FGF, VEGF), and markers of EMT, in part through the post-transcriptional mechanisms described above.
In this review we have examined only a small portion of published scientific data in the field of post-transcriptional regulation of cytokine and growth-factor signaling. New research results will undoubtedly contribute to further expansion of our knowledge of post-transcriptional networks and their complexity. For example, other proteins and miRNAs that directly associate with RNA-RBPs may introduce conformational changes, and alter RBP interactions with mRNAs in the malignant milieu [196–198]. Building a comprehensive cancer interactome of RNA-BPs, in malignant and tumor-associated cells, may provide insight into dysregulated post-transcriptional events. As a result, useful prognostic markers of cancer progression may be realized.
9. Summary
The important roles of AREs and GREs in regulating the dynamic expression of cytokine and growth factors as well as their signaling pathways is increasingly appreciated [199]. Many studies suggest malfunction of ELAVL1 in cancer causes overexpression and stabilization of target mRNAs [47, 200]. In contrast, CELF1 and ZFP36 normally function to limit cellular proliferation, and their inability to function in cancer further promotes uncontrolled cell growth. Thus, the opposing effects of CELF1 and ZFP36 versus ELAVL1 has important implications for the role of post-transcriptional regulation in proliferative diseases such as cancer. Maintaining appropriate balances between RNA-BPs that degrade or stabilize ARE- or GRE-containing mRNAs is a mechanism to ensure appropriate expression of these transcripts and to sustain healthy homeostasis. Understanding the crosstalk between CELF1, ZFP36, ELAVL1 and other RNA-BPs may uncover novel therapeutic strategies to fine-tune the balance in cell growth and cell death pathways involved in human cancer [201, 202]. In the future, more work needs to be done to develop interventions targeting post-transcriptional mechanisms involved in cancer pathogenesis.
Highlights.
Examination of published scientific data in the field of post-transcriptional regulation of cytokine and growth-factor signaling.
AREs and GREs regulate mRNA turnover to coordinate cytokine and growth factor expression.
Three RNA-binding proteins that competitively regulate mRNA stability through ARE and GRE are discussed.
Dysregulation of mRNA degradation contributes to the development and progression of cancer.
Acknowledgments
This work is supported by start-up funds from the Department of Medicine at the University of Minnesota to I.A.V. and National Institutes of Health grants AI072068 and AI096925 to P.R.B. The authors would like to thank Alex Krona for the critical reading and editing of this manuscript. The University of Minnesota Supercomputing Institute provided the access to Ingenuity Pathway Assistant.
Abbreviations
- 3’UTR
untranslated region
- ARE
AU-rich element
- GRE
GU-rich element
- RNA-BP
RNA-binding protein
- ARE-PB
ARE-binding protein
- GRE-BP
GRE-binding protein
- ELAVL1
embryonic lethal, abnormal vision-like 1 (Hu antigen R)
- ZFP36
zinc finger protein 36
- TTP
tristetraprolin
- CELF1
CUGBP-ELAV-like family member 1
- HNRNPD
heterogeneous nuclear ribonucleoprotein D
- KSRP
KH-type splicing regulatory protein
- GAPDH
glyceraldehyde 3-phosphate dehydrogenase
- VEGF
vascular endothelial growth factor
- VEGFR
vascular endothelial growth factor receptor
- GF
growth factor
- GFRs
growth factor receptors
- uPA
urokinase plasminogen activator
- CXCL8
C-X-C motif ligand 8, IL, Interleukin
- PKC
protein kinase C
- MAPK
mitogen-activated protein kinase
- mTOR
mammalian target of rapamycin
- MMP9
matrix metalloproteinase 9
- NFkB
nuclear factor kappa B
- STAT
signal transducer and activator of transcription
- FGFs
fibroblast growth factors
- EGF
epidermal growth factor
- TNFα
tumor necrosis factor-alpha
- ICAM
intercellular adhesion molecule
- VCAM
vascular cell adhesion molecule
- CSF1
colony-stimulating factor one
- DM1
myotonic dystrophy type 1
- EMT
epithelial-to-mesenchymal transitions
Biographies

Irina St. Louis earned her M.D. and Ph.D. degrees from Ural State Medical Academy, Yekaterinburg, Russia. She completed her residency in lab pathology at Russian Medical Academy for Postgraduate Education, Moscow. Dr. St. Louis joined the Department of Microbiology, at University of Minnesota, as a postdoctoral trainee, followed by fellowships at the University of Minnesota Supercomputing Institute and Lymphoma Research Foundation. In 2013, Dr. St. Louis was appointed as an Assistant Professor at the Department of Medicine. The research in her Lab is centered on the area of post-transcriptional gene expression regulation, specifically through messenger RNA degradation. Dr. St. Louis conducts discovery research in several key areas including infectious diseases, immunology, and oncology.

Paul R. Bohjanen, MD, PhD, Professor of Medicine, Division Director of Infectious Diseases & International Medicine, Director of the Center for Infectious Diseases and Microbiology Translational Research (CIDMTR). He graduated from the University of Michigan with an M.D./Ph.D. degrees, and then completed an internal medicine residency and an infectious diseases fellowship in 2000, both at Duke University. Research in his laboratory focuses on the role of mRNA decay in regulating T lymphocyte activation and function. Many genes that are important for cell growth and immune function are turned on at precise times and turned off at precise times. Dysregulated expression of many of these genes, including proto-oncogenes and cytokine genes, occurs in disease states such as cancer, autoimmunity, or immunodeficiency.
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 citable 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 cited
- 1.Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer. 2008;8(11):887–99. doi: 10.1038/nrc2507. [DOI] [PubMed] [Google Scholar]
- 2.West NR, McCuaig S, Franchini F, Powrie F. Emerging cytokine networks in colorectal cancer. Nat Rev Immunol. 2015;15(10):615–29. doi: 10.1038/nri3896. [DOI] [PubMed] [Google Scholar]
- 3.Friedel CC, Dolken L, Ruzsics Z, Koszinowski UH, Zimmer R. Conserved principles of mammalian transcriptional regulation revealed by RNA half-life. Nucleic Acids Res. 2009;37(17):e115. doi: 10.1093/nar/gkp542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Palanisamy V, Jakymiw A, Van Tubergen EA, D'Silva NJ, Kirkwood KL. Control of cytokine mRNA expression by RNA-binding proteins and microRNAs. J Dent Res. 2012;91(7):651–8. doi: 10.1177/0022034512437372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Saharinen P, Eklund L, Pulkki K, Bono P, Alitalo K. VEGF and angiopoietin signaling in tumor angiogenesis and metastasis. Trends Mol Med. 2011;17(7):347–62. doi: 10.1016/j.molmed.2011.01.015. [DOI] [PubMed] [Google Scholar]
- 6.Duan J, Shi J, Ge X, Dolken L, Moy W, He D, Shi S, Sanders AR, Ross J, Gejman PV. Genome-wide survey of interindividual differences of RNA stability in human lymphoblastoid cell lines. Scientific reports. 2013;3:1318. doi: 10.1038/srep01318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Vlasova-St Louis I, Bohjanen PR. Post-transcriptional regulation of cytokine signaling by AU-rich and GU-rich elements. J Interferon Cytokine Res. 2014;34(4):233–41. doi: 10.1089/jir.2013.0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Aparicio LA, Abella V, Valladares M, Figueroa A. Posttranscriptional regulation by RNA-binding proteins during epithelial-to-mesenchymal transition. Cell Mol Life Sci. 2013;70(23):4463–77. doi: 10.1007/s00018-013-1379-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Blackinton JG, Keene JD. Post-transcriptional RNA regulons affecting cell cycle and proliferation. Seminars in cell & developmental biology. 2014 doi: 10.1016/j.semcdb.2014.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Morris AR, Mukherjee N, Keene JD. Systematic analysis of posttranscriptional gene expression. Wiley Interdiscip Rev Syst Biol Med. 2010;2(2):162–80. doi: 10.1002/wsbm.54. [DOI] [PubMed] [Google Scholar]
- 11.Howley BV, Hussey GS, Link LA, Howe PH. Translational regulation of inhibin betaA by TGFbeta via the RNA-binding protein hnRNP E1 enhances the invasiveness of epithelial-to-mesenchymal transitioned cells. Oncogene. 2016;35(13):1725–35. doi: 10.1038/onc.2015.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Qi MY, Wang ZZ, Zhang Z, Shao Q, Zeng A, Li XQ, Li WQ, Wang C, Tian FJ, Li Q, Zou J, Qin YW, Brewer G, Huang S, Jing Q. AU-rich-element-dependent translation repression requires the cooperation of tristetraprolin and RCK/P54. Mol Cell Biol. 2012;32(5):913–28. doi: 10.1128/MCB.05340-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Adjibade P, St-Sauveur VG, Quevillon Huberdeau M, Fournier MJ, Savard A, Coudert L, Khandjian EW, Mazroui R. Sorafenib, a multikinase inhibitor, induces formation of stress granules in hepatocarcinoma cells. Oncotarget. 2015;6(41):43927–43. doi: 10.18632/oncotarget.5980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Adjibade P, Mazroui R. Control of mRNA turnover: implication of cytoplasmic RNA granules. Seminars in cell & developmental biology. 2014;34:15–23. doi: 10.1016/j.semcdb.2014.05.013. [DOI] [PubMed] [Google Scholar]
- 15.Keene JD. RNA regulons: coordination of post-transcriptional events. Nat Rev Genet. 2007;8(7):533–43. doi: 10.1038/nrg2111. [DOI] [PubMed] [Google Scholar]
- 16.von Roretz C, Gallouzi IE. Decoding ARE-mediated decay: is microRNA part of the equation? J Cell Biol. 2008;181(2):189–94. doi: 10.1083/jcb.200712054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Vasudevan S. Posttranscriptional upregulation by microRNAs. Wiley Interdiscip Rev RNA. 2012;3(3):311–30. doi: 10.1002/wrna.121. [DOI] [PubMed] [Google Scholar]
- 18.Di Leva G, Garofalo M, Croce CM. MicroRNAs in cancer. Annual review of pathology. 2014;9:287–314. doi: 10.1146/annurev-pathol-012513-104715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tan JY, Marques AC. The miRNA-mediated cross-talk between transcripts provides a novel layer of posttranscriptional regulation. Adv Genet. 2014;85:149–99. doi: 10.1016/B978-0-12-800271-1.00003-2. [DOI] [PubMed] [Google Scholar]
- 20.Beisang D, Bohjanen PR. Perspectives on the ARE as it turns 25 years old. Wiley Interdiscip Rev RNA. 2012;3(5):719–31. doi: 10.1002/wrna.1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chen CY, Shyu AB. AU-rich elements: characterization and importance in mRNA degradation. Trends Biochem Sci. 1995;20(11):465–70. doi: 10.1016/s0968-0004(00)89102-1. [DOI] [PubMed] [Google Scholar]
- 22.Bakheet T, Williams BR, Khabar KS. ARED 2.0: an update of AU-rich element mRNA database. Nucleic Acids Res. 2003;31(1):421–3. doi: 10.1093/nar/gkg023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bakheet T, Williams BR, Khabar KS. ARED 3.0: the large and diverse AU-rich transcriptome. Nucleic Acids Res. 2006;34(Database issue):D111–4. doi: 10.1093/nar/gkj052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Halees AS, El-Badrawi R, Khabar KS. ARED Organism: expansion of ARED reveals AU-rich element cluster variations between human and mouse. Nucleic Acids Res. 2008;36(Database issue):D137–40. doi: 10.1093/nar/gkm959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bakheet T, Frevel M, Williams BR, Greer W, Khabar KS. ARED: human AU-rich element-containing mRNA database reveals an unexpectedly diverse functional repertoire of encoded proteins. Nucleic Acids Res. 2001;29(1):246–54. doi: 10.1093/nar/29.1.246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Taylor GA, Carballo E, Lee DM, Lai WS, Thompson MJ, Patel DD, Schenkman DI, Gilkeson GS, Broxmeyer HE, Haynes BF, Blackshear PJ. A pathogenetic role for TNF alpha in the syndrome of cachexia, arthritis, and autoimmunity resulting from tristetraprolin (TTP) deficiency. Immunity. 1996;4(5):445–54. doi: 10.1016/s1074-7613(00)80411-2. [DOI] [PubMed] [Google Scholar]
- 27.Chen TM, Hsu CH, Tsai SJ, Sun HS. AUF1 p42 isoform selectively controls both steady-state and PGE2-induced FGF9 mRNA decay. Nucleic Acids Res. 2010;38(22):8061–71. doi: 10.1093/nar/gkq717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Suswam EA, Nabors LB, Huang Y, Yang X, King PH. IL-1beta induces stabilization of IL-8 mRNA in malignant breast cancer cells via the 3' untranslated region: Involvement of divergent RNA-binding factors HuR, KSRP and TIAR. Int J Cancer. 2005;113(6):911–9. doi: 10.1002/ijc.20675. [DOI] [PubMed] [Google Scholar]
- 29.White MR, Khan MM, Deredge D, Ross CR, Quintyn R, Zucconi BE, Wysocki VH, Wintrode PL, Wilson GM, Garcin ED. A dimer interface mutation in glyceraldehyde 3-phosphate dehydrogenase regulates its binding to AU-rich RNA. J Biol Chem. 2015;290(7):4129. doi: 10.1074/jbc.A114.618165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Raghavan A, Robison RL, McNabb J, Miller CR, Williams DA, Bohjanen PR. HuA and tristetraprolin are induced following T cell activation and display distinct but overlapping RNA binding specificities. J Biol Chem. 2001;276(51):47958–65. doi: 10.1074/jbc.M109511200. [DOI] [PubMed] [Google Scholar]
- 31.Anderson P. Post-transcriptional regulons coordinate the initiation and resolution of inflammation. Nat Rev Immunol. 2010;10(1):24–35. doi: 10.1038/nri2685. [DOI] [PubMed] [Google Scholar]
- 32.Ivanov P, Anderson P. Post-transcriptional regulatory networks in immunity. Immunol Rev. 2013;253(1):253–72. doi: 10.1111/imr.12051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Khabar KS. Rapid transit in the immune cells: the role of mRNA turnover regulation. J Leukoc Biol. 2007;81(6):1335–44. doi: 10.1189/jlb.0207109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Schott J, Stoecklin G. Networks controlling mRNA decay in the immune system. Wiley Interdiscip Rev RNA. 2010;1(3):432–56. doi: 10.1002/wrna.13. [DOI] [PubMed] [Google Scholar]
- 35.Villarino AV, Katzman SD, Gallo E, Miller O, Jiang S, McManus MT, Abbas AK. Posttranscriptional silencing of effector cytokine mRNA underlies the anergic phenotype of self-reactive T cells. Immunity. 2011;34(1):50–60. doi: 10.1016/j.immuni.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Raghavan A, Bohjanen PR. Microarray-based analyses of mRNA decay in the regulation of mammalian gene expression. Brief Funct Genomic Proteomic. 2004;3(2):112–24. doi: 10.1093/bfgp/3.2.112. [DOI] [PubMed] [Google Scholar]
- 37.Hau HH, Walsh RJ, Ogilvie RL, Williams DA, Reilly CS, Bohjanen PR. Tristetraprolin recruits functional mRNA decay complexes to ARE sequences. Journal of cellular biochemistry. 2007;100(6):1477–92. doi: 10.1002/jcb.21130. [DOI] [PubMed] [Google Scholar]
- 38.Brooks SA, Blackshear PJ. Tristetraprolin (TTP): Interactions with mRNA and proteins, and current thoughts on mechanisms of action. Biochimica et Biophysica Acta. 2013 doi: 10.1016/j.bbagrm.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Paschoud S, Dogar AM, Kuntz C, Grisoni-Neupert B, Richman L, Kuhn LC. Destabilization of interleukin-6 mRNA requires a putative RNA stem-loop structure, an AU-rich element, and the RNA-binding protein AUF1. Mol Cell Biol. 2006;26(22):8228–41. doi: 10.1128/MCB.01155-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Malathi K, Paranjape JM, Bulanova E, Shim M, Guenther-Johnson JM, Faber PW, Eling TE, Williams BR, Silverman RH. A transcriptional signaling pathway in the IFN system mediated by 2'-5'-oligoadenylate activation of RNase L. Proc Natl Acad Sci U S A. 2005;102(41):14533–8. doi: 10.1073/pnas.0507551102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fialcowitz EJ, Brewer BY, Keenan BP, Wilson GM. A hairpin-like structure within an AU-rich mRNA-destabilizing element regulates trans-factor binding selectivity and mRNA decay kinetics. J Biol Chem. 2005;280(23):22406–17. doi: 10.1074/jbc.M500618200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Al-Haj L, Blackshear PJ, Khabar KS. Regulation of p21/CIP1/WAF-1 mediated cell-cycle arrest by RNase L and tristetraprolin, and involvement of AU-rich elements. Nucleic Acids Res. 2012;40(16):7739–52. doi: 10.1093/nar/gks545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vlasova IA, McNabb J, Raghavan A, Reilly C, Williams DA, Bohjanen KA, Bohjanen PR. Coordinate stabilization of growth-regulatory transcripts in T cell malignancies. Genomics. 2005;86(2):159–71. doi: 10.1016/j.ygeno.2005.04.013. [DOI] [PubMed] [Google Scholar]
- 44.Chu PC, Kulp SK, Chen CS. Insulin-like growth factor-I receptor is suppressed through transcriptional repression and mRNA destabilization by a novel energy restriction-mimetic agent. Carcinogenesis. 2013;34(12):2694–705. doi: 10.1093/carcin/bgt251. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 45.Hitti E, Khabar KS. Sequence variations affecting AU-rich element function and disease. Frontiers in bioscience. 2012;17:1846–60. doi: 10.2741/4023. [DOI] [PubMed] [Google Scholar]
- 46.Al-Ahmadi W, Al-Ghamdi M, Al-Haj L, Al-Saif M, Khabar KS. Alternative polyadenylation variants of the RNA binding protein, HuR: abundance, role of AU-rich elements and auto-Regulation. Nucleic Acids Res. 2009;37(11):3612–24. doi: 10.1093/nar/gkp223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hitti E, Bakheet T, Al-Souhibani N, Moghrabi W, Al-Yahya S, Al-Ghamdi M, Al-Saif M, Shoukri MM, Lanczky A, Grepin R, Gyorffy B, Pages G, Khabar KS. Systematic Analysis of AU-Rich Element Expression in Cancer Reveals Common Functional Clusters Regulated by Key RNA-Binding Proteins. Cancer Res. 2016 doi: 10.1158/0008-5472.CAN-15-3110. [DOI] [PubMed] [Google Scholar]
- 48.Raghavan A, Ogilvie RL, Reilly C, Abelson ML, Raghavan S, Vasdewani J, Krathwohl M, Bohjanen PR. Genome-wide analysis of mRNA decay in resting and activated primary human T lymphocytes. Nucleic Acids Res. 2002;30(24):5529–38. doi: 10.1093/nar/gkf682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.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 biology. 2014;15(1):R12. doi: 10.1186/gb-2014-15-1-r12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Atasoy U, Curry SL, Lopez de Silanes I, Shyu AB, Casolaro V, Gorospe M, Stellato C. Regulation of eotaxin gene expression by TNF-alpha and IL-4 through mRNA stabilization: involvement of the RNA-binding protein HuR. J Immunol. 2003;171(8):4369–78. doi: 10.4049/jimmunol.171.8.4369. [DOI] [PubMed] [Google Scholar]
- 51.Calaluce R, Gubin MM, Davis JW, Magee JD, Chen J, Kuwano Y, Gorospe M, Atasoy U. The RNA binding protein HuR differentially regulates unique subsets of mRNAs in estrogen receptor negative and estrogen receptor positive breast cancer. BMC Cancer. 2010;10:126. doi: 10.1186/1471-2407-10-126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Soler DM, Ghosh A, Chen F, Shneider BL. A single element in the 3'UTR of the apical sodium-dependent bile acid transporter controls both stabilization and destabilization of mRNA. Biochem J. 2014;462(3):547–53. doi: 10.1042/BJ20140070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kotta-Loizou I, Giaginis C, Theocharis S. Clinical significance of HuR expression in human malignancy. Medical oncology. 2014;31(9):161. doi: 10.1007/s12032-014-0161-y. [DOI] [PubMed] [Google Scholar]
- 54.Sanduja S, Blanco FF, Young LE, Kaza V, Dixon DA. The role of tristetraprolin in cancer and inflammation. Frontiers in bioscience. 2012;17:174–88. doi: 10.2741/3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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. 2008;68(3):674–82. doi: 10.1158/0008-5472.CAN-07-2751. [DOI] [PubMed] [Google Scholar]
- 56.Basso K, Saito M, Sumazin P, Margolin AA, Wang K, Lim WK, Kitagawa Y, Schneider C, Alvarez MJ, Califano A, Dalla-Favera R. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood. 2010;115(5):975–84. doi: 10.1182/blood-2009-06-227017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Stumpo DJ, Broxmeyer HE, Ward T, Cooper S, Hangoc G, Chung YJ, Shelley WC, Richfield EK, Ray MK, Yoder MC, Aplan PD, Blackshear PJ. Targeted disruption of Zfp36l2, encoding a CCCH tandem zinc finger RNA-binding protein, results in defective hematopoiesis. Blood. 2009;114(12):2401–10. doi: 10.1182/blood-2009-04-214619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Al-Souhibani N, Al-Ghamdi M, Al-Ahmadi W, Khabar KS. Posttranscriptional control of the chemokine receptor CXCR4 expression in cancer cells. Carcinogenesis. 2014;35(9):1983–92. doi: 10.1093/carcin/bgu080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Upadhyay R, Sanduja S, Kaza V, Dixon DA. Genetic polymorphisms in RNA binding proteins contribute to breast cancer survival. Int J Cancer. 2013;132(3):E128–38. doi: 10.1002/ijc.27789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Griseri P, Bourcier C, Hieblot C, Essafi-Benkhadir K, Chamorey E, Touriol C, Pages G. A synonymous polymorphism of the Tristetraprolin (TTP) gene, an AU-rich mRNA-binding protein, affects translation efficiency and response to Herceptin treatment in breast cancer patients. Human molecular genetics. 2011;20(23):4556–68. doi: 10.1093/hmg/ddr390. [DOI] [PubMed] [Google Scholar]
- 61.Lopez de Silanes I, Fan J, Yang X, Zonderman AB, Potapova O, Pizer ES, Gorospe M. Role of the RNA-binding protein HuR in colon carcinogenesis. Oncogene. 2003;22(46):7146–54. doi: 10.1038/sj.onc.1206862. [DOI] [PubMed] [Google Scholar]
- 62.Lauriola L, Granone P, Ramella S, Lanza P, Ranelletti FO. Expression of the RNA-binding protein HuR and its clinical significance in human stage I and II lung adenocarcinoma. Histology and histopathology. 2012;27(5):617–26. doi: 10.14670/HH-27.617. [DOI] [PubMed] [Google Scholar]
- 63.Sun DP, Lin CY, Tian YF, Chen LT, Lin LC, Lee SW, Hsing CH, Lee HH, Shiue YL, Huang HY, Li CF, Liang PI. Clinicopathological significance of HuR expression in gallbladder carcinoma: with special emphasis on the implications of its nuclear and cytoplasmic expression. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2013;34(5):3059–69. doi: 10.1007/s13277-013-0872-2. [DOI] [PubMed] [Google Scholar]
- 64.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. 2011;9(5):648–59. doi: 10.1158/1541-7786.MCR-10-0325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Leijon H, Salmenkivi K, Heiskanen I, Hagstrom J, Louhimo J, Heikkila P, Ristimaki A, Paavonen T, Metso S, Maenpaa H, Haglund C, Arola J. HuR in pheochromocytomas and paragangliomas - overexpression in verified malignant tumors. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica. 2016;124(9):757–63. doi: 10.1111/apm.12571. [DOI] [PubMed] [Google Scholar]
- 66.Kim GY, Lim SJ, Kim YW. Expression of HuR, COX-2, and survivin in lung cancers; cytoplasmic HuR stabilizes cyclooxygenase-2 in squamous cell carcinomas. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2011;24(10):1336–47. doi: 10.1038/modpathol.2011.90. [DOI] [PubMed] [Google Scholar]
- 67.Nabors LB, Gillespie GY, Harkins L, King PH. HuR, a RNA stability factor, is expressed in malignant brain tumors and binds to adenine- and uridine-rich elements within the 3' untranslated regions of cytokine and angiogenic factor mRNAs. Cancer Res. 2001;61(5):2154–61. [PubMed] [Google Scholar]
- 68.Dixon DA, Tolley ND, King PH, Nabors LB, McIntyre TM, Zimmerman GA, Prescott SM. Altered expression of the mRNA stability factor HuR promotes cyclooxygenase-2 expression in colon cancer cells. J Clin Invest. 2001;108(11):1657–65. doi: 10.1172/JCI12973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.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. 2003;63(14):4181–7. [PubMed] [Google Scholar]
- 70.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. 2009;29(16):4341–51. doi: 10.1128/MCB.00210-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Filippova N, Yang X, Nabors LB. Growth factor dependent regulation of centrosome function and genomic instability by HuR. Biomolecules. 2015;5(1):263–81. doi: 10.3390/biom5010263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Doller A, Winkler C, Azrilian I, Schulz S, Hartmann S, Pfeilschifter J, Eberhardt W. High-constitutive HuR phosphorylation at Ser 318 by PKC{delta} propagates tumor relevant functions in colon carcinoma cells. Carcinogenesis. 2011;32(5):676–85. doi: 10.1093/carcin/bgr024. [DOI] [PubMed] [Google Scholar]
- 73.Goldberg-Cohen I, Furneauxb H, Levy AP. A 40-bp RNA element that mediates stabilization of vascular endothelial growth factor mRNA by HuR. J Biol Chem. 2002;277(16):13635–40. doi: 10.1074/jbc.M108703200. [DOI] [PubMed] [Google Scholar]
- 74.Gabai VL, Meng L, Kim G, Mills TA, Benjamin IJ, Sherman MY. Heat shock transcription factor Hsf1 is involved in tumor progression via regulation of hypoxia-inducible factor 1 and RNA-binding protein HuR. Mol Cell Biol. 2012;32(5):929–40. doi: 10.1128/MCB.05921-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhang J, Modi Y, Yarovinsky T, Yu J, Collinge M, Kyriakides T, Zhu Y, Sessa WC, Pardi R, Bender JR. Macrophage beta2 integrin-mediated, HuR-dependent stabilization of angiogenic factor-encoding mRNAs in inflammatory angiogenesis. Am J Pathol. 2012;180(4):1751–60. doi: 10.1016/j.ajpath.2011.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Amadio M, Bucolo C, Leggio GM, Drago F, Govoni S, Pascale A. The PKCbeta/HuR/VEGF pathway in diabetic retinopathy. Biochemical pharmacology. 2010;80(8):1230–7. doi: 10.1016/j.bcp.2010.06.033. [DOI] [PubMed] [Google Scholar]
- 77.Roche E, Lascombe I, Bittard H, Mougin C, Fauconnet S. The PPARbeta agonist L-165041 promotes VEGF mRNA stabilization in HPV18-harboring HeLa cells through a receptor-independent mechanism. Cell Signal. 2014;26(2):433–43. doi: 10.1016/j.cellsig.2013.10.006. [DOI] [PubMed] [Google Scholar]
- 78.Galban S, Kuwano Y, Pullmann R, Jr, Martindale JL, Kim HH, Lal A, Abdelmohsen K, Yang X, Dang Y, Liu JO, Lewis SM, Holcik M, Gorospe M. RNA-binding proteins HuR and PTB promote the translation of hypoxia-inducible factor 1alpha. Mol Cell Biol. 2008;28(1):93–107. doi: 10.1128/MCB.00973-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Blanco FF, Jimbo M, Wulfkuhle J, Gallagher I, Deng J, Enyenihi L, Meisner-Kober N, Londin E, Rigoutsos I, Sawicki JA, Risbud MV, Witkiewicz AK, McCue PA, Jiang W, Rui H, Yeo CJ, Petricoin E, Winter JM, Brody JR. The mRNA-binding protein HuR promotes hypoxia-induced chemoresistance through posttranscriptional regulation of the proto-oncogene PIM1 in pancreatic cancer cells. Oncogene. 2015 doi: 10.1038/onc.2015.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Cheng YC, Liou JP, Kuo CC, Lai WY, Shih KH, Chang CY, Pan WY, Tseng JT, Chang JY. MPT0B098, a novel microtubule inhibitor that destabilizes the hypoxia-inducible factor-1alpha mRNA through decreasing nuclear-cytoplasmic translocation of RNA-binding protein HuR. Molecular cancer therapeutics. 2013;12(7):1202–12. doi: 10.1158/1535-7163.MCT-12-0778. [DOI] [PubMed] [Google Scholar]
- 81.Basu A, Datta D, Zurakowski D, Pal S. Altered VEGF mRNA stability following treatments with immunosuppressive agents: implications for cancer development. Journal of Biological Chemistry. 2010;285(33):25196–202. doi: 10.1074/jbc.M110.119446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Govindaraju S, Lee BS. Adaptive and maladaptive expression of the mRNA regulatory protein HuR. World journal of biological chemistry. 2013;4(4):111–8. doi: 10.4331/wjbc.v4.i4.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Heinonen M, Bono P, Narko K, Chang SH, Lundin J, Joensuu H, Furneaux H, Hla T, Haglund C, Ristimaki A. Cytoplasmic HuR expression is a prognostic factor in invasive ductal breast carcinoma. Cancer Res. 2005;65(6):2157–61. doi: 10.1158/0008-5472.CAN-04-3765. [DOI] [PubMed] [Google Scholar]
- 84.Denkert C, Weichert W, Winzer KJ, Muller BM, Noske A, Niesporek S, Kristiansen G, Guski H, Dietel M, Hauptmann S. Expression of the ELAV-like protein HuR is associated with higher tumor grade and increased cyclooxygenase-2 expression in human breast carcinoma. Clin Cancer Res. 2004;10(16):5580–6. doi: 10.1158/1078-0432.CCR-04-0070. [DOI] [PubMed] [Google Scholar]
- 85.Mazan-Mamczarz K, Hagner PR, Corl S, Srikantan S, Wood WH, Becker KG, Gorospe M, Keene JD, Levenson AS, Gartenhaus RB. Post-transcriptional gene regulation by HuR promotes a more tumorigenic phenotype. Oncogene. 2008;27(47):6151–63. doi: 10.1038/onc.2008.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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. 2015;34(22):2934–42. doi: 10.1038/onc.2014.225. [DOI] [PubMed] [Google Scholar]
- 87.Gubin MM, Calaluce R, Davis JW, Magee JD, Strouse CS, Shaw DP, Ma L, Brown A, Hoffman T, Rold TL, Atasoy U. Overexpression of the RNA binding protein HuR impairs tumor growth in triple negative breast cancer associated with deficient angiogenesis. Cell Cycle. 2010;9(16):3337–46. doi: 10.4161/cc.9.16.12711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Wang J, Li D, Wang B, Wu Y. Predictive and prognostic significance of cytoplasmic expression of ELAV-like protein HuR in invasive breast cancer treated with neoadjuvant chemotherapy. Breast Cancer Res Treat. 2013;141(2):213–24. doi: 10.1007/s10549-013-2679-7. [DOI] [PubMed] [Google Scholar]
- 89.Danilin S, Sourbier C, Thomas L, Lindner V, Rothhut S, Dormoy V, Helwig JJ, Jacqmin D, Lang H, Massfelder T. Role of the RNA-binding protein HuR in human renal cell carcinoma. Carcinogenesis. 2010;31(6):1018–26. doi: 10.1093/carcin/bgq052. [DOI] [PubMed] [Google Scholar]
- 90.Costantino CL, Witkiewicz AK, Kuwano Y, Cozzitorto JA, Kennedy EP, Dasgupta A, Keen JC, Yeo CJ, Gorospe M, Brody JR. The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR Up-regulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase. Cancer Res. 2009;69(11):4567–72. doi: 10.1158/0008-5472.CAN-09-0371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kang MJ, Ryu BK, Lee MG, Han J, Lee JH, Ha TK, Byun DS, Chae KS, Lee BH, Chun HS, Lee KY, Kim HJ, Chi SG. NF-kappaB activates transcription of the RNA-binding factor HuR, via PI3K-AKT signaling, to promote gastric tumorigenesis. Gastroenterology. 2008;135(6):2030–42. 2042 e1–3. doi: 10.1053/j.gastro.2008.08.009. [DOI] [PubMed] [Google Scholar]
- 92.Papadaki O, Milatos S, Grammenoudi S, Mukherjee N, Keene JD, Kontoyiannis DL. Control of thymic T cell maturation, deletion and egress by the RNA-binding protein HuR. J Immunol. 2009;182(11):6779–88. doi: 10.4049/jimmunol.0900377. [DOI] [PubMed] [Google Scholar]
- 93.Yiakouvaki A, Dimitriou M, Karakasiliotis I, Eftychi C, Theocharis S, Kontoyiannis DL. Myeloid cell expression of the RNA-binding protein HuR protects mice from pathologic inflammation and colorectal carcinogenesis. J Clin Invest. 2012;122(1):48–61. doi: 10.1172/JCI45021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Johnson BA, Blackwell TK. Multiple tristetraprolin sequence domains required to induce apoptosis and modulate responses to TNFalpha through distinct pathways. Oncogene. 2002;21(27):4237–46. doi: 10.1038/sj.onc.1205526. [DOI] [PubMed] [Google Scholar]
- 95.Carrick DM, Blackshear PJ. Comparative expression of tristetraprolin (TTP) family member transcripts in normal human tissues and cancer cell lines. Archives of biochemistry and biophysics. 2007;462(2):278–85. doi: 10.1016/j.abb.2007.04.011. [DOI] [PubMed] [Google Scholar]
- 96.Ogilvie RL, Abelson M, Hau HH, Vlasova I, Blackshear PJ, Bohjanen PR. Tristetraprolin down-regulates IL-2 gene expression through AU-rich element-mediated mRNA decay. Journal of Immunology. 2005;174(2):953–61. doi: 10.4049/jimmunol.174.2.953. [DOI] [PubMed] [Google Scholar]
- 97.Essafi-Benkhadir K, Onesto C, Stebe E, Moroni C, Pages G. Tristetraprolin inhibits Ras-dependent tumor vascularization by inducing vascular endothelial growth factor mRNA degradation. Mol Biol Cell. 2007;18(11):4648–58. doi: 10.1091/mbc.E07-06-0570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schaljo B, Kratochvill F, Gratz N, Sadzak I, Sauer I, Hammer M, Vogl C, Strobl B, Muller M, Blackshear PJ, Poli V, Lang R, Murray PJ, Kovarik P. Tristetraprolin is required for full anti-inflammatory response of murine macrophages to IL-10. Journal of Immunology. 2009;183(2):1197–206. doi: 10.4049/jimmunol.0803883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lee HH, Yoon NA, Vo MT, Kim CW, Woo JM, Cha HJ, Cho YW, Lee BJ, Cho WJ, Park JW. Tristetraprolin down-regulates IL-17 through mRNA destabilization. FEBS Lett. 2012;586(1):41–6. doi: 10.1016/j.febslet.2011.11.021. [DOI] [PubMed] [Google Scholar]
- 100.Qian X, Ning H, Zhang J, Hoft DF, Stumpo DJ, Blackshear PJ, Liu J. Posttranscriptional regulation of IL-23 expression by IFN-gamma through tristetraprolin. J Immunol. 2011;186(11):6454–64. doi: 10.4049/jimmunol.1002672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Milke L, Schulz K, Weigert A, Sha W, Schmid T, Brune B. Depletion of tristetraprolin in breast cancer cells increases interleukin-16 expression and promotes tumor infiltration with monocytes/macrophages. Carcinogenesis. 2013;34(4):850–7. doi: 10.1093/carcin/bgs387. [DOI] [PubMed] [Google Scholar]
- 102.Molle C, Zhang T, Ysebrant de Lendonck L, Gueydan C, Andrianne M, Sherer F, Van Simaeys G, Blackshear PJ, Leo O, Goriely S. Tristetraprolin regulation of interleukin 23 mRNA stability prevents a spontaneous inflammatory disease. J Exp Med. 2013;210(9):1675–84. doi: 10.1084/jem.20120707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ogilvie RL, Sternjohn JR, Rattenbacher B, Vlasova IA, Williams DA, Hau HH, Blackshear PJ, Bohjanen PR. Tristetraprolin mediates interferon-gamma mRNA decay. J Biol Chem. 2009;284(17):11216–23. doi: 10.1074/jbc.M901229200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lee HH, Yang SS, Vo MT, Cho WJ, Lee BJ, Leem SH, Lee SH, Cha HJ, Park JW. Tristetraprolin down-regulates IL-23 expression in colon cancer cells. Mol Cells. 2013;36(6):571–6. doi: 10.1007/s10059-013-0268-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gebeshuber CA, Zatloukal K, Martinez J. miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep. 2009;10(4):400–5. doi: 10.1038/embor.2009.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Fahling M, Persson AB, Klinger B, Benko E, Steege A, Kasim M, Patzak A, Persson PB, Wolf G, Bluthgen N, Mrowka R. Multilevel regulation of HIF-1 signaling by TTP. Mol Biol Cell. 2012;23(20):4129–41. doi: 10.1091/mbc.E11-11-0949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Brennan-Laun SE, Li XL, Ezelle HJ, Venkataraman T, Blackshear PJ, Wilson GM, Hassel BA. RNase L attenuates mitogen-stimulated gene expression via transcriptional and post-transcriptional mechanisms to limit the proliferative response. J Biol Chem. 2014;289(48):33629–43. doi: 10.1074/jbc.M114.589556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Patial S, Curtis AD, 2nd, Lai WS, Stumpo DJ, Hill GD, Flake GP, Mannie MD, Blackshear PJ. Enhanced stability of tristetraprolin mRNA protects mice against immune-mediated inflammatory pathologies. Proc Natl Acad Sci U S A. 2016;113(7):1865–70. doi: 10.1073/pnas.1519906113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Huang L, Yu Z, Zhang Z, Ma W, Song S, Huang G. Interaction with Pyruvate Kinase M2 Destabilizes Tristetraprolin by Proteasome Degradation and Regulates Cell Proliferation in Breast Cancer. Scientific reports. 2016;6:22449. doi: 10.1038/srep22449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ross CR, Brennan-Laun SE, Wilson GM. Tristetraprolin: roles in cancer and senescence. Ageing Res Rev. 2012;11(4):473–84. doi: 10.1016/j.arr.2012.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Van Tubergen E, Vander Broek R, Lee J, Wolf G, Carey T, Bradford C, Prince M, Kirkwood KL, D'Silva NJ. Tristetraprolin regulates interleukin-6, which is correlated with tumor progression in patients with head and neck squamous cell carcinoma. Cancer. 2011;117(12):2677–89. doi: 10.1002/cncr.25859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Al-Souhibani N, Al-Ahmadi W, Hesketh JE, Blackshear PJ, Khabar KS. The RNA-binding zinc-finger protein tristetraprolin regulates AU-rich mRNAs involved in breast cancer-related processes. Oncogene. 2010;29(29):4205–15. doi: 10.1038/onc.2010.168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Brennan SE, Kuwano Y, Alkharouf N, Blackshear PJ, Gorospe M, Wilson GM. The mRNA-destabilizing protein tristetraprolin is suppressed in many cancers, altering tumorigenic phenotypes and patient prognosis. Cancer Res. 2009;69(12):5168–76. doi: 10.1158/0008-5472.CAN-08-4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bourcier C, Griseri P, Grepin R, Bertolotto C, Mazure N, Pages G. Constitutive ERK activity induces downregulation of tristetraprolin, a major protein controlling interleukin8/CXCL8 mRNA stability in melanoma cells. American journal of physiology. Cell physiology. 2011;301(3):C609–18. doi: 10.1152/ajpcell.00506.2010. [DOI] [PubMed] [Google Scholar]
- 115.Young LE, Sanduja S, Bemis-Standoli K, Pena EA, Price RL, Dixon DA. The mRNA binding proteins HuR and tristetraprolin regulate cyclooxygenase 2 expression during colon carcinogenesis. Gastroenterology. 2009;136(5):1669–79. doi: 10.1053/j.gastro.2009.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Mahat DB, Brennan-Laun SE, Fialcowitz-White EJ, Kishor A, Ross CR, Pozharskaya T, Rawn JD, Blackshear PJ, Hassel BA, Wilson GM. Coordinated expression of tristetraprolin post-transcriptionally attenuates mitogenic induction of the oncogenic Ser/Thr kinase Pim-1. PLoS One. 2012;7(3):e33194. doi: 10.1371/journal.pone.0033194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Bolognani F, Gallani AI, Sokol L, Baskin DS, Meisner-Kober N. mRNA stability alterations mediated by HuR are necessary to sustain the fast growth of glioma cells. J Neurooncol. 2012;106(3):531–42. doi: 10.1007/s11060-011-0707-1. [DOI] [PubMed] [Google Scholar]
- 118.Al-Ahmadi W, Al-Ghamdi M, Al-Souhibani N, Khabar KS. miR-29a inhibition normalizes HuR over-expression and aberrant AU-rich mRNA stability in invasive cancer. Journal of Pathology. 2013;230(1):28–38. doi: 10.1002/path.4178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.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. 2013;113(2):195–205. doi: 10.1007/s11060-013-1112-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Jimbo M, Blanco FF, Huang YH, Telonis AG, Screnci BA, Cosma GL, Alexeev V, Gonye GE, Yeo CJ, Sawicki JA, Winter JM, Brody JR. Targeting the mRNA-binding protein HuR impairs malignant characteristics of pancreatic ductal adenocarcinoma cells. Oncotarget. 2015 doi: 10.18632/oncotarget.4743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yoon JH, Abdelmohsen K, Srikantan S, Guo R, Yang X, Martindale JL, Gorospe M. Tyrosine phosphorylation of HuR by JAK3 triggers dissociation and degradation of HuR target mRNAs. Nucleic Acids Res. 2014;42(2):1196–208. doi: 10.1093/nar/gkt903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sobolewski C, Sanduja S, Blanco FF, Hu L, Dixon DA. Histone Deacetylase Inhibitors Activate Tristetraprolin Expression through Induction of Early Growth Response Protein 1 (EGR1) in Colorectal Cancer Cells. Biomolecules. 2015;5(3):2035–55. doi: 10.3390/biom5032035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Vlasova IA, Bohjanen PR. Posttranscriptional regulation of gene networks by GU-rich elements and CELF proteins. RNA Biol. 2008;5(4):201–7. doi: 10.4161/rna.7056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Vlasova IA, Tahoe NM, Fan D, Larsson O, Rattenbacher B, Sternjohn JR, Vasdewani J, Karypis G, Reilly CS, Bitterman PB, Bohjanen PR. Conserved GU-rich elements mediate mRNA decay by binding to CUG-binding protein 1. Mol Cell. 2008;29(2):263–70. doi: 10.1016/j.molcel.2007.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Halees AS, Hitti E, Al-Saif M, Mahmoud L, Vlasova-St Louis IA, Beisang DJ, Bohjanen PR, Khabar K. Global assessment of GU-rich regulatory content and function in the human transcriptome. RNA Biol. 2011;8(4):681–91. doi: 10.4161/rna.8.4.16283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Rattenbacher B, Beisang D, Wiesner DL, Jeschke JC, von Hohenberg M, St Louis-Vlasova IA, Bohjanen PR. Analysis of CUGBP1 targets identifies GU-repeat sequences that mediate rapid mRNA decay. Mol Cell Biol. 2010;30(16):3970–80. doi: 10.1128/MCB.00624-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Rolle K, Piwecka M, Belter A, Wawrzyniak D, Jeleniewicz J, Barciszewska MZ, Barciszewski J. The Sequence and Structure Determine the Function of Mature Human miRNAs. PLoS One. 2016;11(3):e0151246. doi: 10.1371/journal.pone.0151246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Katoh T, Hojo H, Suzuki T. Destabilization of microRNAs in human cells by 3' deadenylation mediated by PARN and CUGBP1. Nucleic Acids Res. 2015;43(15):7521–34. doi: 10.1093/nar/gkv669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Vlasova-St Louis I, Bohjanen PR. Coordinate regulation of mRNA decay networks by GU-rich elements and CELF1. Curr Opin Genet Dev. 2011;21(4):444–51. doi: 10.1016/j.gde.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Beisang D, Rattenbacher B, Vlasova-St Louis IA, Bohjanen PR. Regulation of CUG-binding Protein 1 (CUGBP1) Binding to Target Transcripts upon T Cell Activation. Journal of Biological Chemistry. 2012;287(2):950–960. doi: 10.1074/jbc.M111.291658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lee JE, Lee JY, Wilusz J, Tian B, Wilusz CJ. Systematic analysis of cis-elements in unstable mRNAs demonstrates that CUGBP1 is a key regulator of mRNA decay in muscle cells. PLoS One. 2010;5(6):e11201. doi: 10.1371/journal.pone.0011201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Blech-Hermoni Y, Dasgupta T, Coram RJ, Ladd AN. Identification of Targets of CUG-BP, Elav-Like Family Member 1 (CELF1) Regulation in Embryonic Heart Muscle. PLoS One. 2016;11(2):e0149061. doi: 10.1371/journal.pone.0149061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Vlasova-St Louis I, Bohjanen PR. Post-transcriptional Regulation of Cytokine Signaling During Inflammatory Responses. In: Goldstrohm AC, Menon KMJ, editors. Post-transcriptional Mechanisms in Endocrine Regulation. Springer; Switzerland: 2016. pp. 55–70. [Google Scholar]
- 134.Beisang D, Bohjanen P, Vlasova-St Louis I. CELF1, a Multifunctional Regulator of Posttranscriptional Networks. InTech Chapter. 2012;8(Binding Protein):181–206. [Google Scholar]
- 135.Liu Y, Huang H, Yuan B, Luo T, Li J, Qin X. Suppression of CUGBP1 inhibits growth of hepatocellular carcinoma cells. Clin Invest Med. 2014;37(1):E10–8. doi: 10.25011/cim.v37i1.20864. [DOI] [PubMed] [Google Scholar]
- 136.Wang X, Wang H, Ji F, Zhao S, Fang X. Lentivirus-mediated knockdown of CUGBP1 suppresses gastric cancer cell proliferation in vitro. Appl Biochem Biotechnol. 2014;173(6):1529–36. doi: 10.1007/s12010-014-0937-8. [DOI] [PubMed] [Google Scholar]
- 137.Bohjanen PR, Moua ML, Guo L, Taye A, Vlasova-St Louis IA. Altered CELF1 binding to target transcripts in malignant T cells. RNA. 2015 doi: 10.1261/rna.049940.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Zhao J, Zhao Y, Xuan Y, Jiao W, Qiu T, Wang Z, Luo Y. Prognostic impact of CUG-binding protein 1 expression and vascular invasion after radical surgery for stage IB nonsmall cell lung cancer. Indian journal of cancer. 2015;52(Suppl 2):e125–9. doi: 10.4103/0019-509X.172509. [DOI] [PubMed] [Google Scholar]
- 139.Gao C, Yu Z, Liu S, Xin H, Li X. Overexpression of CUGBP1 is associated with the progression of non-small cell lung cancer. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2015;36(6):4583–9. doi: 10.1007/s13277-015-3103-1. [DOI] [PubMed] [Google Scholar]
- 140.Vlasova-St Louis I, Dickson AM, Bohjanen PR, Wilusz CJ. CELFish ways to modulate mRNA decay. Biochimica Et Biophysica Acta-Gene Regulatory Mechanisms. 2013;1829(6–7):695–707. doi: 10.1016/j.bbagrm.2013.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Le Tonqueze O, Gschloessl B, Legagneux V, Paillard L, Audic Y. Identification of CELF1 RNA targets by CLIP-seq in human HeLa cells. Genomics data. 2016;8:97–103. doi: 10.1016/j.gdata.2016.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Timchenko LT, Timchenko NA, Caskey CT, Roberts R. Novel proteins with binding specificity for DNA CTG repeats and RNA CUG repeats: implications for myotonic dystrophy. Human molecular genetics. 1996;5(1):115–21. doi: 10.1093/hmg/5.1.115. [DOI] [PubMed] [Google Scholar]
- 143.Mueller CM, Hilbert JE, Martens W, Thornton CA, Moxley RT, 3rd, Greene MH. Hypothesis: neoplasms in myotonic dystrophy. Cancer causes & control : CCC. 2009;20(10):2009–20. doi: 10.1007/s10552-009-9395-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Gadalla SM, Lund M, Pfeiffer RM, Gortz S, Mueller CM, Moxley RT, 3rd, Kristinsson SY, Bjorkholm M, Shebl FM, Hilbert JE, Landgren O, Wohlfahrt J, Melbye M, Greene MH. Cancer risk among patients with myotonic muscular dystrophy. JAMA : the journal of the American Medical Association. 2011;306(22):2480–6. doi: 10.1001/jama.2011.1796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Win AK, Perattur PG, Pulido JS, Pulido CM, Lindor NM. Increased cancer risks in myotonic dystrophy. Mayo Clinic proceedings. 2012;87(2):130–5. doi: 10.1016/j.mayocp.2011.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Guerzoni C, Bardini M, Mariani SA, Ferrari-Amorotti G, Neviani P, Panno ML, Zhang Y, Martinez R, Perrotti D, Calabretta B. Inducible activation of CEBPB, a gene negatively regulated by BCR/ABL, inhibits proliferation and promotes differentiation of BCR/ABL-expressing cells. Blood. 2006;107(10):4080–9. doi: 10.1182/blood-2005-08-3181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Jin J, Iakova P, Jiang Y, Lewis K, Sullivan E, Jawanmardi N, Donehower L, Timchenko L, Timchenko NA. Transcriptional and translational regulation of C/EBPbeta-HDAC1 protein complexes controls different levels of p53, SIRT1, and PGC1alpha proteins at the early and late stages of liver cancer. J Biol Chem. 2013;288(20):14451–62. doi: 10.1074/jbc.M113.460840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Arnal-Estape A, Tarragona M, Morales M, Guiu M, Nadal C, Massague J, Gomis RR. HER2 silences tumor suppression in breast cancer cells by switching expression of C/EBPss isoforms. Cancer Res. 2010;70(23):9927–36. doi: 10.1158/0008-5472.CAN-10-0869. [DOI] [PubMed] [Google Scholar]
- 149.Liu L, Ouyang M, Rao JN, Zou T, Xiao L, Chung HK, Wu J, Donahue JM, Gorospe M, Wang JY. Competition between RNA-binding proteins CELF1 and HuR modulates MYC translation and intestinal epithelium renewal. Mol Biol Cell. 2015;26(10):1797–810. doi: 10.1091/mbc.E14-11-1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lebedeva S, Jens M, Theil K, Schwanhausser B, Selbach M, Landthaler M, Rajewsky N. Transcriptome-wide analysis of regulatory interactions of the RNA-binding protein HuR. Mol Cell. 2011;43(3):340–52. doi: 10.1016/j.molcel.2011.06.008. [DOI] [PubMed] [Google Scholar]
- 151.Schulz S, Doller A, Pendini NR, Wilce JA, Pfeilschifter J, Eberhardt W. Domain-specific phosphomimetic mutation allows dissection of different protein kinase C (PKC) isotype-triggered activities of the RNA binding protein HuR. Cell Signal. 2013;25(12):2485–95. doi: 10.1016/j.cellsig.2013.08.003. [DOI] [PubMed] [Google Scholar]
- 152.Mukherjee N, Corcoran DL, Nusbaum JD, Reid DW, Georgiev S, Hafner M, Ascano M, Jr, Tuschl T, Ohler U, Keene JD. Integrative regulatory mapping indicates that the RNA-binding protein HuR couples pre-mRNA processing and mRNA stability. Mol Cell. 2011;43(3):327–39. doi: 10.1016/j.molcel.2011.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kuyumcu-Martinez NM, Wang GS, Cooper TA. Increased steady-state levels of CUGBP1 in myotonic dystrophy 1 are due to PKC-mediated hyperphosphorylation. Mol Cell. 2007;28(1):68–78. doi: 10.1016/j.molcel.2007.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Erkinheimo TL, Lassus H, Sivula A, Sengupta S, Furneaux H, Hla T, Haglund C, Butzow R, Ristimaki A. Cytoplasmic HuR expression correlates with poor outcome and with cyclooxygenase 2 expression in serous ovarian carcinoma. Cancer Res. 2003;63(22):7591–4. [PubMed] [Google Scholar]
- 155.Wang J, Wang B, Bi J, Zhang C. Cytoplasmic HuR expression correlates with angiogenesis, lymphangiogenesis, and poor outcome in lung cancer. Medical oncology. 2011;28(Suppl 1):S577–85. doi: 10.1007/s12032-010-9734-6. [DOI] [PubMed] [Google Scholar]
- 156.Zhu Z, Wang B, Bi J, Zhang C, Guo Y, Chu H, Liang X, Zhong C, Wang J. Cytoplasmic HuR expression correlates with P-gp, HER-2 positivity, and poor outcome in breast cancer. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2013;34(4):2299–308. doi: 10.1007/s13277-013-0774-3. [DOI] [PubMed] [Google Scholar]
- 157.Vlasova-St Louis I, Bohjanen PR. Feedback Regulation of Kinase Signaling Pathways by AREs and GREs. Cells. 2016;5(1) doi: 10.3390/cells5010004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.von Roretz C, Macri AM, Gallouzi IE. Transportin 2 regulates apoptosis through the RNA-binding protein HuR. J Biol Chem. 2011;286(29):25983–91. doi: 10.1074/jbc.M110.216184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Simone LE, Keene JD. Mechanisms coordinating ELAV/Hu mRNA regulons. Curr Opin Genet Dev. 2013;23(1):35–43. doi: 10.1016/j.gde.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473(7347):298–307. doi: 10.1038/nature10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Yu TX, Rao JN, Zou T, Liu L, Xiao L, Ouyang M, Cao S, Gorospe M, Wang JY. Competitive binding of CUGBP1 and HuR to occludin mRNA controls its translation and modulates epithelial barrier function. Mol Biol Cell. 2013;24(2):85–99. doi: 10.1091/mbc.E12-07-0531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Yu TX, Gu BL, Yan JK, Zhu J, Yan WH, Chen J, Qian LX, Cai W. CUGBP1 and HuR regulate E-cadherin translation by altering recruitment of E-cadherin mRNA to processing bodies and modulate epithelial barrier function. American journal of physiology. Cell physiology. 2016;310(1):C54–65. doi: 10.1152/ajpcell.00112.2015. [DOI] [PubMed] [Google Scholar]
- 163.Yang H, Rao JN, Wang JY. Posttranscriptional Regulation of Intestinal Epithelial Tight Junction Barrier by RNA-binding Proteins and microRNAs. Tissue Barriers. 2014;2(1):e28320. doi: 10.4161/tisb.28320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bai D, Gao Q, Li C, Ge L, Gao Y, Wang H. A conserved TGFbeta1/HuR feedback circuit regulates the fibrogenic response in fibroblasts. Cell Signal. 2012;24(7):1426–32. doi: 10.1016/j.cellsig.2012.03.003. [DOI] [PubMed] [Google Scholar]
- 165.Ge J, Chang N, Zhao Z, Tian L, Duan X, Yang L, Li L. Essential Roles of RNA-binding Protein HuR in Activation of Hepatic Stellate Cells Induced by Transforming Growth Factor-beta1. Scientific reports. 2016;6:22141. doi: 10.1038/srep22141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Kanies CL, Smith JJ, Kis C, Schmidt C, Levy S, Khabar KS, Morrow J, Deane N, Dixon DA, Beauchamp RD. Oncogenic Ras and transforming growth factor-beta synergistically regulate AU-rich element-containing mRNAs during epithelial to mesenchymal transition. Mol Cancer Res. 2008;6(7):1124–36. doi: 10.1158/1541-7786.MCR-07-2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Han Y, Wang X, Wang B, Jiang G. The progress of angiogenic factors in the development of leukemias. Intractable & rare diseases research. 2016;5(1):6–16. doi: 10.5582/irdr.2015.01048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Van Tubergen EA, Banerjee R, Liu M, Vander Broek R, Light E, Kuo S, Feinberg SE, Willis AL, Wolf G, Carey T, Bradford C, Prince M, Worden FP, Kirkwood KL, D'Silva NJ. Inactivation or loss of TTP promotes invasion in head and neck cancer via transcript stabilization and secretion of MMP9, MMP2, and IL-6. Clin Cancer Res. 2013;19(5):1169–79. doi: 10.1158/1078-0432.CCR-12-2927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell. 2010;17(2):135–47. doi: 10.1016/j.ccr.2009.12.041. [DOI] [PubMed] [Google Scholar]
- 170.Yuan Z, Sanders AJ, Ye L, Wang Y, Jiang WG. Knockdown of human antigen R reduces the growth and invasion of breast cancer cells in vitro and affects expression of cyclin D1 and MMP-9. Oncology reports. 2011;26(1):237–45. doi: 10.3892/or.2011.1271. [DOI] [PubMed] [Google Scholar]
- 171.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(20):7177–88. doi: 10.1128/MCB.23.20.7177-7188.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Fuxe J, Karlsson MC. TGF-beta-induced epithelial-mesenchymal transition: a link between cancer and inflammation. Semin Cancer Biol. 2012;22(5–6):455–61. doi: 10.1016/j.semcancer.2012.05.004. [DOI] [PubMed] [Google Scholar]
- 173.Singh M, Martinez AR, Govindaraju S, Lee BS. HuR inhibits apoptosis by amplifying Akt signaling through a positive feedback loop. J Cell Physiol. 2013;228(1):182–9. doi: 10.1002/jcp.24120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Talwar S, Balasubramanian S, Sundaramurthy S, House R, Wilusz CJ, Kuppuswamy D, D'Silva N, Gillespie MB, Hill EG, Palanisamy V. Overexpression of RNA-binding protein CELF1 prevents apoptosis and destabilizes pro-apoptotic mRNAs in oral cancer cells. RNA Biol. 2013;10(2):277–86. doi: 10.4161/rna.23315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Durie D, Lewis SM, Liwak U, Kisilewicz M, Gorospe M, Holcik M. RNA-binding protein HuR mediates cytoprotection through stimulation of XIAP translation. Oncogene. 2011;30(12):1460–9. doi: 10.1038/onc.2010.527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Durie D, Hatzoglou M, Chakraborty P, Holcik M. HuR controls mitochondrial morphology through the regulation of Bcl translation. Translation (Austin) 2013;1(1) doi: 10.4161/trla.23980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Liang PI, Li WM, Wang YH, Wu TF, Wu WR, Liao AC, Shen KH, Wei YC, Hsing CH, Shiue YL, Huang HY, Hsu HP, Chen LT, Lin CY, Tai C, Lin CM, Li CF. HuR cytoplasmic expression is associated with increased cyclin A expression and poor outcome with upper urinary tract urothelial carcinoma. BMC Cancer. 2012;12:611. doi: 10.1186/1471-2407-12-611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Dong AQ, Kong MJ, Ma ZY, Qian JF, Xu XH. Down-regulation of IGF-IR using small, interfering, hairpin RNA (siRNA) inhibits growth of human lung cancer cell line A549 in vitro and in nude mice. Cell biology international. 2007;31(5):500–7. doi: 10.1016/j.cellbi.2006.11.017. [DOI] [PubMed] [Google Scholar]
- 179.Zhou Y, Chang R, Ji W, Wang N, Qi M, Xu Y, Guo J, Zhan L. Loss of Scribble Promotes Snail Translation through Translocation of HuR and Enhances Cancer Drug Resistance. J Biol Chem. 2016;291(1):291–302. doi: 10.1074/jbc.M115.693853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.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 beta-catenin in HCC cells. Gastroenterology. 2015;148(2):415–26 e18. doi: 10.1053/j.gastro.2014.10.012. [DOI] [PubMed] [Google Scholar]
- 181.Yoon NA, Jo HG, Lee UH, Park JH, Yoon JE, Ryu J, Kang SS, Min YJ, Ju SA, Seo EH, Huh IY, Lee BJ, Park JW, Cho WJ. Tristetraprolin suppresses the EMT through the down-regulation of Twist1 and Snail1 in cancer cells. Oncotarget. 2016;7(8):8931–43. doi: 10.18632/oncotarget.7094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Luo NA, Qu YQ, Yang GD, Wang T, Li RL, Jia LT, Dong R. Post-transcriptional up-regulation of PDGF-C by HuR in advanced and stressed breast cancer. International journal of molecular sciences. 2014;15(11):20306–20. doi: 10.3390/ijms151120306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.D'Uva G, Bertoni S, Lauriola M, De Carolis S, Pacilli A, D'Anello L, Santini D, Taffurelli M, Ceccarelli C, Yarden Y, Montanaro L, Bonafe M, Storci G. Beta-catenin/HuR post-transcriptional machinery governs cancer stem cell features in response to hypoxia. PLoS One. 2013;8(11):e80742. doi: 10.1371/journal.pone.0080742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Datta S, Biswas R, Novotny M, Pavicic PG, Jr, Herjan T, Mandal P, Hamilton TA. Tristetraprolin regulates CXCL1 (KC) mRNA stability. J Immunol. 2008;180(4):2545–52. doi: 10.4049/jimmunol.180.4.2545. [DOI] [PubMed] [Google Scholar]
- 185.Kratochvill F, Gratz N, Qualls JE, Van De Velde LA, Chi H, Kovarik P, Murray PJ. Tristetraprolin Limits Inflammatory Cytokine Production in Tumor-Associated Macrophages in an mRNA Decay-Independent Manner. Cancer Res. 2015;75(15):3054–64. doi: 10.1158/0008-5472.CAN-15-0205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Blanco FF, Jimbo M, Wulfkuhle J, Gallagher I, Deng J, Enyenihi L, Meisner-Kober N, Londin E, Rigoutsos I, Sawicki JA, Risbud MV, Witkiewicz AK, McCue PA, Jiang W, Rui H, Yeo CJ, Petricoin E, Winter JM, Brody JR. The mRNA-binding protein HuR promotes hypoxia-induced chemoresistance through posttranscriptional regulation of the proto-oncogene PIM1 in pancreatic cancer cells. Oncogene. 2016;35(19):2529–41. doi: 10.1038/onc.2015.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Griseri P, Pages G. Control of pro-angiogenic cytokine mRNA half-life in cancer: the role of AU-rich elements and associated proteins. J Interferon Cytokine Res. 2014;34(4):242–54. doi: 10.1089/jir.2013.0140. [DOI] [PubMed] [Google Scholar]
- 188.Borsig L, Wolf MJ, Roblek M, Lorentzen A, Heikenwalder M. Inflammatory chemokines and metastasis--tracing the accessory. Oncogene. 2014;33(25):3217–24. doi: 10.1038/onc.2013.272. [DOI] [PubMed] [Google Scholar]
- 189.Kim I, Hur J, Jeong S. HuR represses Wnt/beta-catenin-mediated transcriptional activity by promoting cytoplasmic localization of beta-catenin. Biochem Biophys Res Commun. 2015;457(1):65–70. doi: 10.1016/j.bbrc.2014.12.052. [DOI] [PubMed] [Google Scholar]
- 190.Shu Y, Pi F, Sharma A, Rajabi M, Haque F, Shu D, Leggas M, Evers BM, Guo P. Stable RNA nanoparticles as potential new generation drugs for cancer therapy. Advanced drug delivery reviews. 2014;66:74–89. doi: 10.1016/j.addr.2013.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Fan Y, Shen B, Tan M, Mu X, Qin Y, Zhang F, Liu Y. Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating Wnt signaling. FEBS J. 2014;281(7):1750–8. doi: 10.1111/febs.12737. [DOI] [PubMed] [Google Scholar]
- 192.Hodson DJ, Janas ML, Galloway A, Bell SE, Andrews S, Li CM, Pannell R, Siebel CW, MacDonald HR, De Keersmaecker K, Ferrando AA, Grutz G, Turner M. Deletion of the RNA-binding proteins ZFP36L1 and ZFP36L2 leads to perturbed thymic development and T lymphoblastic leukemia. Nat Immunol. 2010;11(8):717–24. doi: 10.1038/ni.1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Kang JG, Amar MJ, Remaley AT, Kwon J, Blackshear PJ, Wang PY, Hwang PM. Zinc finger protein tristetraprolin interacts with CCL3 mRNA and regulates tissue inflammation. J Immunol. 2011;187(5):2696–701. doi: 10.4049/jimmunol.1101149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Sohn BH, Park IY, Lee JJ, Yang SJ, Jang YJ, Park KC, Kim DJ, Lee DC, Sohn HA, Kim TW, Yoo HS, Choi JY, Bae YS, Yeom YI. Functional switching of TGF-beta1 signaling in liver cancer via epigenetic modulation of a single CpG site in TTP promoter. Gastroenterology. 2010;138(5):1898–908. doi: 10.1053/j.gastro.2009.12.044. [DOI] [PubMed] [Google Scholar]
- 195.Donahue JM, Chang ET, Xiao L, Wang PY, Rao JN, Turner DJ, Wang JY, Battafarano RJ. The RNA-binding protein HuR stabilizes survivin mRNA in human oesophageal epithelial cells. The Biochemical journal. 2011;437(1):89–96. doi: 10.1042/BJ20110028. [DOI] [PubMed] [Google Scholar]
- 196.Krishnan N, Titus MA, Thapar R. The prolyl isomerase pin1 regulates mRNA levels of genes with short half-lives by targeting specific RNA binding proteins. PLoS One. 2014;9(1):e85427. doi: 10.1371/journal.pone.0085427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Vantourout P, Willcox C, Turner A, Swanson CM, Haque Y, Sobolev O, Grigoriadis A, Tutt A, Hayday A. Immunological visibility: posttranscriptional regulation of human NKG2D ligands by the EGF receptor pathway. Sci Transl Med. 2014;6(231):231ra49. doi: 10.1126/scitranslmed.3007579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.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. 2016;35(13):1703–15. doi: 10.1038/onc.2015.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Khabar KS. Post-transcriptional control of cytokine gene expression in health and disease. J Interferon Cytokine Res. 2014;34(4):215–9. doi: 10.1089/jir.2013.0151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Khabar KS. Post-transcriptional control during chronic inflammation and cancer: a focus on AU-rich elements. Cellular and Molecular Life Sciences. 2010;67(17):2937–55. doi: 10.1007/s00018-010-0383-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Eberhardt W, Badawi A, Biyanee A, Pfeilschifter J. Cytoskeleton-Dependent Transport as a Potential Target for Interfering with Post-transcriptional HuR mRNA Regulons. Frontiers in pharmacology. 2016;7:251. doi: 10.3389/fphar.2016.00251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Meng M, Wang W, Yan J, Tan J, Liao L, Shi J, Wei C, Xie Y, Jin X, Yang L, Jin Q, Zhu H, Tan W, Yang F, Hou Z. Immunization of stromal cell targeting fibroblast activation protein providing immunotherapy to breast cancer mouse model. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2016;37(8):10317–27. doi: 10.1007/s13277-016-4825-4. [DOI] [PubMed] [Google Scholar]



