Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a key tumor suppressor in the development and progression of different tumor types. Emerging data indicate that small reductions in PTEN protein levels can promote cancer. PTEN protein levels are tightly controlled by a plethora of mechanisms beginning with epigenetic and transcriptional regulation and ending with control of protein synthesis and stability. PTEN messenger RNA (mRNA) is also subject to exquisite regulation by microRNAs, coding and long noncoding RNAs, and RNA-binding proteins. Additionally, PTEN mRNA is markedly influenced by alternative splicing and variable polyadenylation. Herein we provide a synoptic description of the current understanding of the complex regulatory landscape of PTEN mRNA regulation including several specific processes that modulate its stability and expression, in the context of PTEN loss-associated cancers.
Exquisite regulation of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) protein expression is critical to suppress the development and progression of many tumor types. Cellular levels of PTEN protein are governed by a variety of mechanisms, including histone modification, DNA methylation, transcriptional regulation, translational regulation, and posttranslational modifications (Song et al. 2012; Correia et al. 2014; Lee et al. 2018). Interestingly, observations made in mouse models indicate that small changes in PTEN protein levels can have profound consequences on tumor incidence, penetrance, and aggressiveness (Alimonti et al. 2010; Carracedo et al. 2011). Unlike classical models of tumor suppression where genetic perturbations are proposed to lead to allelic losses and consequent gross losses (50% or 100%) of protein expression or activity, the “continuum model” highlights the importance of nongenomic losses of tumor suppressor expression by more subtle mechanisms, such as the regulation by microRNA (miRNA) or other posttranscriptional mechanisms regulating PTEN messenger RNA (mRNA) levels (Berger and Pandolfi 2011).
Clinical observations that brain, prostate, endometrial, and gastric tissues, among others, are sensitive to small nongenetic perturbations in PTEN levels support the notion that PTEN may comply with a “continuum model” of tumor suppression rather than the more classical “two-hit” or haploinsufficiency models (Kwabi-Addo et al. 2001; Trotman et al. 2003; Alimonti et al. 2010; Berger and Pandolfi 2011). In this review, we focus on PTEN transcript regulation. We present and discuss progress in the understanding of PTEN mRNA regulation by various mechanisms including miRNA, competing endogenous RNA (ceRNA), alternative splicing, and use of alternative polyadenylation sites on PTEN transcripts, as well as the impact of N6-methyladenosine (m6A) modification.
M. icroRNA and PTEN
miRNA are small noncoding RNA molecules of ∼19–24 nucleotides in length that mediate gene silencing and play important roles in a wide range of fundamental biological processes and a variety of diverse diseases. miRNA are an essential component of the miRNA-induced silencing complex (miRISC), a multiprotein RNA complex that contains members of the Argonaute (Ago) family of proteins (Jonas and Izaurralde 2015). The principal function of miRNA is to guide the miRISC to specific RNA targets through partial Watson–Crick base pairing, typically on the 3′ untranslated region (UTR) of protein-coding and noncoding transcripts (Chandradoss et al. 2015). miRISC binding to a 3′UTR leads to gene silencing through translation repression and mRNA decay (Grimson et al. 2007; Bartel 2009; Kehl et al. 2017). Because miRNA can repress expression of most genes without completely silencing them, they are considered an important class of factors with an ability to fine-tune nearly all cellular processes.
Interest in regulation of the PTEN mRNA transcript by miRNA is extensive, with currently over 1500 publications that describe interactions of PTEN mRNA with one or more miRNA. While the landscape of miRNA regulation is complex and thought to be context-dependent, there are numerous studies that have identified specific high confidence miRNA:PTEN interactions that also present biological relevance. The pleiotropic function of PTEN in many cancers is mirrored by the miRNA-mediated regulation, which have been found to target and silence PTEN expression in numerous cancers including non-small-cell lung cancer (NSCLC), colorectal carcinoma, ovarian cancer, triple-negative breast cancers, prostate cancer, and melanoma (for review, see Bermúdez Brito et al. 2015). In each case, silencing of PTEN by miRNA leads to more aggressive forms of the disease. For example, miR-221 and miR-222 are highly expressed in NSCLC and hepatocellular carcinoma, and promote tumorigenesis by targeting PTEN (Garofalo et al. 2009). In the same fashion, PTEN was repressed by aberrant overexpression of the miR-106b∼25 cluster in human prostate cancer, leading to more aggressive forms of prostate adenocarcinoma in mouse models (Poliseno et al. 2010a).
The first validated PTEN miRNA recognition element (MRE) was that of miR-21 (Ji et al. 2007; Meng et al. 2007), which was shown to bind to the PTEN 3′UTR (Ji et al. 2007; Meng et al. 2007) and is generally regarded as a potent bona fide PTEN targeting miRNA in several malignant and nonmalignant contexts (Feng and Tsao 2016). PTEN has been reported to interact with many other miRNA families, and it is widely accepted that expression of PTEN is heavily regulated through the function of miRNA. For instance, members of miR-9, -17, -19, -21, -23, -25, -26, -33, -103, -130, -136, -144, -154, -181, -214, -216, -217, -221, -222, and -506 gene families have all been implicated in fine-tuning PTEN expression in cancer and other contexts (detailed in Table 1).
Table 1.
miRNA gene family | miRNA examined | Cell model | Tissue type | Biological evidence | Luciferase reporter assay | MRE mutated | References |
---|---|---|---|---|---|---|---|
miR-9 | miR-9 | TU212, Hep-2 | Laryngocarcinoma | Yes | Yes | Yes | Lu et al. 2016 |
miR-17 | miR-17 | DU145 | Prostate cancer | Yes | Yes | Yes | Poliseno et al. 2010a |
miR-17-5p | NIH3T3 | Mouse fibroblast | Yes | Yes | Yes | Xiao et al. 2008 | |
miR-18a-5p | U2OS | Osteosarcoma | Yes | NA | NA | Fei et al. 2018 | |
miR-20a | DU145/HEK 293T/HUVEC/HEK 293 | Prostate cancer/SV40 large T antigen model/endothelial/model line | Yes/yes/yes/yes | NA/yes/yes/yes | NA/yes/yes/yes | Poliseno et al. 2010b; Zhang et al. 2015; Wang et al. 2017a; Jiang et al. 2018 | |
miR-93 | MCF-7, MDA-MB-231 | Breast cancer | Yes | Yes | Yes | Li et al. 2017a | |
miR-106a | SGC7901/PC-3 | Gastric cancer/prostate cancer | Yes/yes | Yes/yes | Yes/NA | Fang et al. 2013/Lu et al. 2019 | |
miR-106a-3p | HT29 | Colorectal adenocarcinoma | Yes | Yes | Yes | Qin et al. 2018 | |
miR-106b | MCF-7 and MDA-MB-231 | Breast cancer | Yes | Yes | Yes | Li et al. 2017a | |
miR-19 | miR-19 | DU145 | Prostate cancer | Yes | Yes | Yes | Poliseno et al. 2010a |
miR-19a | NCTC1469 cells and HEP1–6/HOS | Macrophage-like and hepatocellular carcinoma/osteosarcoma | Yes/yes | Yes/yes | NA/yes | Dou et al. 2015; Zhao et al. 2017 | |
miR-19a-3p | U2OS | Osteosarcoma | Yes | Yes | Yes | Zhang et al. 2019 | |
miR-19b | DU145/H9C2/MDA-MB-231 | Prostate cancer/rat cardiomyocyte/breast cancer | Yes/yes/yes | NA/NA/yes | NA/NA/yes | Poliseno et al. 2010b; Xu et al. 2016; Li et al. 2017b | |
miR-21 | miR-21 | SK-HEP-1 and SNU-182/VSMC/A549 | Hepatocellular carcinoma/vascular smooth muscles/non-small lung cancer | Yes/yes/yes | Yes/NA/NA | NA/NA/NA | Meng et al. 2006, 2007; Ji et al. 2007; |
miR-22 | miR-22 | DU145 | Prostate cancer | Yes | Yes | Yes | Poliseno et al. 2010a |
miR-23 | miR-23a-3p | HEK 293 | Model | Yes | Yes | NA | Lozano-Bartolomé et al. 2018 |
miR-23b-3p | A-498 | Kidney carcinoma | Yes | Yes | NA | Zaman et al. 2012 | |
miR-25 | miR-25 | DU145 | Prostate cancer | Yes | Yes | Yes | Poliseno et al. 2010a |
miR-92a | U2OS/U14/A549 | Osteosarcoma/cervical cancer/adenocarcinoma | Yes/yes/yes | Yes/yes/yes | Yes/yes/yes | Ren et al. 2016; Li et al. 2017b; Xiao et al. 2017 | |
miR-26 | miR-26a | HEK 293 | Model | Yes | Yes | Yes | Huse et al. 2009 |
miR-33 | miR-33a | HEK 293 | Model | Yes | Yes | NA | Ling et al. 2013 |
miR-103 | miR-103 | UMUC2 and 5637 | Bladder cancer | Yes | Yes | Yes | Yu et al. 2019 |
miR-103a | HEK 293/HEK 293 | Model/model | Yes/yes | Yes/yes | Yes/yes | Geng et al. 2014; Hsu et al. 2017 | |
miR-107 | EJ cells | Bladder cancer | Yes | Yes | Yes | Chi et al. 2018; Yu et al. 2019 | |
miR-130 | miR-130b | HEK 293T /HCCLM3 and HepG2 | SV40 large T antigen model/hepatocellular carcinoma | Yes/yes | Yes/yes | Yes/yes | Chang et al. 2016; Miao et al. 2017 |
miR-136 | miR-136 | U343 | Glioblastoma | Yes | Yes | Yes | Lee et al. 2010 |
miR-144 | miR-144 | U343 | Glioblastoma | Yes | Yes | Yes | Lee et al. 2010 |
miR-154 | miR-494 | Huh-7/INS-1 | Hepatocellular carcinoma/rat insulinoma | Yes/yes | NA/yes | NA/yes | He et al. 2017; Pollutri et al. 2018 |
miR-494-3p | SMMC-7721 and HCCLM3 | HPV-related endocervical adenocarcinoma | Yes | Yes | Yes | Lin et al. 2018 | |
miR-181 | miR-181-5p | HEK 293 | Model | Yes | Yes | NA | Lozano-Bartolomé et al. 2018 |
miR-193 | miR-193a | 786-O cells | Renal cell carcinoma | Yes | Yes | Yes | Liu et al. 2017 |
miR-214 | miR-214 | A2780CP and HIOSE-80 | Ovarian cancer | Yes | Yes | Yes | Yang et al. 2008 |
miR-216 | miR-216a | Mouse glomerular mesangial cells | Normal kidney | Yes | Yes | Yes | Kato et al. 2009 |
miR-216a+miR-217 | PLC/-PRF/5 | Hepatocellular carcinoma | Yes | Yes | Yes | Xia et al. 2013 | |
miR-217 | miR-217 | mouse glomerular mesangial cells/HEK 293T | Normal kidney/SV40 large T antigen model | Yes/yes | Yes/yes | Yes/yes | Kato et al. 2009; Nie et al. 2018 |
miR-221 | miR-222 | OVCAR-3 | Ovarian cancer | Yes | Yes | Yes | Zhou et al. 2012; Gong et al. 2018 |
miR-302 | miR-302 | DU145 | Prostate cancer | Yes | Yes | Yes | Poliseno et al. 2010a |
miR-506 | miR-510-3p | NCI-H441 and PC-9 | Non-small lung cancer | Yes | Yes | Yes | Yu et al. 2019 |
Validation ranges from having any biological evidence such as western blots and qPCR data, to analyses of the isolated fragment of the 3′UTR in a luciferase reporter assay, which may have the miRNA recognition element (MRE) mutated to further demonstrate loss of repression.
(NA) not applicable.
An ongoing challenge in the study of miRNA biology is the correct identification of miRNA:mRNA interactions; this is especially arduous because each miRNA has numerous mRNA targets and vice versa. Prediction of an MRE within a transcript typically begins with the use of one of many prediction algorithms. The diverse biological features that govern miRNA binding to an MRE are differentially weighed within these algorithms, further to being informed by experimental data (Liu et al. 2013a,b; Peterson et al. 2014). Subsequent validation of miRNA:mRNA interactions range from further in silico associations to in-depth in vitro studies and may include (1) reporter construct assays to show a direct miRNA/transcript association, (2) validation with a mutated MRE, and (3) miRNA-inhibitor-mediated derepression of expression. Advanced techniques such as high-throughput sequencing of RNAs isolated by cross-linking immunoprecipitation (HITS-CLIP) or photoactivatable ribonucleoside-enhanced CLIP (PAR-CLIP) have experimentally identified functional miRNA:mRNA interaction maps (Chi et al. 2009; Hafner et al. 2010). Although these emerging techniques are complex and biochemically challenging, they provide great promise for high-throughput simultaneous discovery of many miRNA:mRNA interactions. Indeed, a recent study by Zarringhalam and colleagues has analyzed multiple datasets from PAR-CLIP experiments in conjunction with RNA-Seq data to evaluate PTEN miRNA networks and their putative biological functions (Zarringhalam et al. 2017).
Competing Endogenous RNA and PTEN
It has been hypothesized that RNA transcripts have the potential to regulate the expression of other RNAs in trans. Such transcripts have been termed ceRNA because of their ability to effectively bind and sequester specific miRNAs, which prevents them from binding to other target RNAs (Poliseno et al. 2010b; Cesana et al. 2011; Salmena et al. 2011). The ceRNA hypothesis posits that as the levels of one RNA transcript that contains a given MRE increases, the levels of another mRNA that contains that same MRE also increases in a dose-dependent manner (Seitz 2009; Salmena et al. 2011; Ala et al. 2013). Despite recognition of the ceRNA hypothesis, a full understanding of ceRNA is still in its infancy and its generalized function, physiological ramifications, and widespread acceptance remain contentious (Bosson et al. 2014; Denzler et al. 2014, 2016). Nevertheless, close to 50 publications collectively describe a large network of coding and noncoding ceRNA that regulate PTEN expression (Table 2). This collection of articles suggest that the PTEN “ceRNome” can influence PTEN expression and, as a consequence, perturbations in this network may have effects on PTEN function and tumorigenesis (Fig. 1).
Table 2.
ceRNA | miRNA implicated | Disease/condition | References |
---|---|---|---|
PTENP1 | miR-17, miR-19, miR-21, miR-214, and miR-26 | Prostate cancer and others | Poliseno et al. 2010b |
FER 14 | miR-106a, miR-18a-5p | Gastric cancer, osteosarcoma | Xia et al. 2015; Yue et al. 2015 |
MEG3 | miR19a | Glioma | Qin et al. 2017 |
HOTAIR | miR19a | Cardiac hypertrophy | Lai et al. 2017 |
SLC25A5-AS1 | miR19a | Gastric cancer | Li et al. 2019 |
Linc-USP16 | miR21 | Breast cancer | Li et al. 2017a |
Gas5 | miR-222-3p and miR-21 | Papillary thyroid carcinoma/breast cancer | Zhang et al. 2013, 2018b; Gong et al. 2018 |
NEAT1 | miR9 | Bladder cancer | Lu et al. 2016; Xie et al. 2018 |
TP73-AS1 | miR103 | Colon cancer | Jia et al. 2019; Yu et al. 2019 |
LINC00702 | miR-510 | Non-small-cell lung cancer (NSCLC) | Yu et al. 2019 |
ORLNC1 | miR-296 | Osteogenesis and adipogenesis | Kakizaki et al. 2017; Yang et al. 2018 |
The first ceRNA interaction was reported between PTEN and PTEN pseudogene 1 (PTENP1) (Poliseno et al. 2010b). The PTENP1 locus was first described in 1998 in two concurrent studies (Dahia et al. 1998; Whang et al. 1998). PTENP1, localized at 9p13.3 is a processed pseudogene that is highly homologous to PTEN containing only 18 mismatches throughout the coding sequence and a missense mutation of the initiator methionine codon that prevents translation, thus sharing several common MREs with the PTEN mRNA (Fujii et al. 1999). Like PTEN, PTENP1 was demonstrated to have tumor-suppressive features including growth suppression on loss of expression in some cancers (Poliseno et al. 2010b).
Consistent with the ceRNA hypothesis, it was observed that PTEN mRNA expression is directly proportional to PTENP1 transcript expression in diverse settings. This was posited to be a consequence of PTEN and PTENP1 transcripts simultaneously controlled by several miRNA at their shared MREs (Poliseno et al. 2010b; Ioffe et al. 2012; Kovalenko et al. 2013, 2018; Yu et al. 2014). Within highly homologous regions, MREs for miR-17, miR-19, miR-21, and miR-214 families have been found, with miR-19b and miR-20a shown to directly down-regulate PTENP1 (Poliseno et al. 2010b). Subsequent studies expanded the list of miRNAs with direct effects on both PTEN and PTENP1, to include miR-26a and miR-193a-3p (Poliseno et al. 2010b; Chen et al. 2015; Gao et al. 2017; Li et al. 2017b; Qian et al. 2017; Yu et al. 2017; Yndestad et al. 2018). By mutating the MRE sequences of these miRNA families within the PTENP1 gene, degradation of PTENP1 was impaired, the effect on PTENP1 expression was derepressed, and PTEN protein levels decreased (Poliseno et al. 2010b; Yu et al. 2014). To date, PTENP1 and PTEN interactions have been reported to influence several tumor types including NSCLC, prostate cancer, melanoma, endometrial cancer, acute myeloid leukemia, clear-cell renal carcinoma, bladder cancer, hepatocellular carcinoma, gastric cancer, oral squamous cell carcinoma, and breast cancer (Marsit et al. 2005; Poliseno et al. 2010b, 2011; Ioffe et al. 2012; Wang et al. 2012; Yu et al. 2014, 2017; Chen et al. 2015; Guo et al. 2016; Gao et al. 2017; Li et al. 2017b; Qian et al. 2017; Yndestad et al. 2017, 2018; Zhang et al. 2017; Shi et al. 2018).
Beyond the PTENP1 pseudogene, a number of additional long noncoding RNA (lncRNAs) have also been reported to regulate PTEN through the ceRNA mechanism. By exploring the association between miR-106a and PTEN, and the data demonstrating miR-106a MRE on FER1L4, it was postulated that the ceRNA network of the FER1L4 lncRNA includes PTEN (Xia et al. 2014a). This possibility was supported by data in gastric cancer where FER1L4 was down-regulated and that its level corresponded with that of PTEN mRNA. Further experimental data showing that FER1L4 down-regulation decreased the abundances of PTEN mRNA supported these findings (Xia et al. 2015; Yue et al. 2015). In a related study, miR-18a-5p was found to mediate a ceRNA interaction between FER1L4 and PTEN in osteosarcoma (Fei et al. 2018). miR-19a was shown to mediate a direct and reciprocal ceRNA interaction between PTEN and MEG3 in glioma (Qin et al. 2017); HOTAIR and PTEN in cardiac hypertrophy (Lai et al. 2017) and SLC25A5-AS1 and PTEN in gastric cancer (Lai et al. 2017; Li et al. 2019). miR-21 mediates a ceRNA interaction between Linc-USP16 and PTEN in hepatocellular carcinoma where Linc-USP16 overexpression also resulted in increased PTEN expression (Zhang et al. 2010; Yang et al. 2013; Sui et al. 2017). Finally, lncRNAs including Gas5, LINC00702, NEAT1, RP11-79H23.3 and TP73-AS1, and ORLNC1 have also been reported to regulate PTEN through the ceRNA mechanism (Xia et al. 2014a; Kakizaki et al. 2017; Lai et al. 2017; Qin et al. 2017; Sui et al. 2017; Chi et al. 2018; Xie et al. 2018; Yang et al. 2018; Jia et al. 2019; Li et al. 2019; Yu et al. 2019).
In addition to lncRNA acting as a ceRNA, several protein-coding genes have also been reported to act as a ceRNA for PTEN. For example, expression of VCAN was found to modulate the expression of PTEN and RB1 (Lee et al. 2010). Both PTEN and VCAN were down-regulated in response to miR-144 and miR-136 expression, supporting a possible interaction through a ceRNA mechanism (Lee et al. 2010). PTEN was later reported to be coregulated with the DKK1 transcript, as both are inhibited by miR-217, -33a, -33b, -103a, -93, and -106a, through a proposed ceRNA interaction (Ling et al. 2013). Similarly, both CASC2 and PTEN mRNA contain miR-21 MREs, and are posited to be coregulated through a ceRNA regulatory mechanism (Meng et al. 2007; Zhang et al. 2010; Feng et al. 2017). Finally, variations in SERINC1, VAPA, CNOT6L, and ZEB2 transcripts were reported to impact PTEN expression levels through ceRNA (Karreth et al. 2011; Tay et al. 2011).
Large numbers of other genes including TNRC6B, RB1, TP53, NRAS, KLF6, HIF1A, HIAT1, CTBP2, and TNKS2 and other very large RNA networks have been identified as PTEN ceRNA through bioinformatic approaches; however, many such computational efforts lack any extensive experimental validation (Sumazin et al. 2011; Zarringhalam et al. 2017; Chiu et al. 2018). Overall, there is an overwhelming abundance of data suggesting that both coding and noncoding transcripts can act as PTEN ceRNAs; however, many studies fail to perform experimental validation that will be necessary to identify bona fide PTEN ceRNA and ceRNA networks.
PTEN Regulation by RNA-Binding Proteins
RNA-binding proteins (RBPs) are critical regulators of transcriptional and posttranscriptional gene expression in all eukaryotic organisms. Unlike other regulatory modalities affecting PTEN expression such as miRNA, there is limited evidence on the role of RBPs in PTEN biology. The most extensively studied PTEN:RBP interaction is with the RNA-binding motif protein 38 (RBM38), an RBP and a target of the P53 family, which has broad roles in transcriptional and posttranscriptional regulation. RBM38 directly interacts with, and increases the stability of, PTEN mRNA by binding to the AU-rich elements within the 3′UTR in multiple breast cancer cell lines (Zhou et al. 2017). Subsequent work in a Rbm38 knockout mouse confirmed that by binding to and stabilizing Pten mRNA, Rbm38 was integral to Pten tumor suppression and that loss of Rbm38 synergizes with mutant p53 to promote lymphomagenesis (Zhang et al. 2018a). Related studies indicate that RBM38 may be occluding miRNA binding to the PTEN-3′UTR and thereby stabilizing PTEN mRNA, as shown previously for the TP53 transcript (Léveillé et al. 2011). RBM38 may also affect PTEN mRNA splicing, similar to its impact on red blood cell (Heinicke et al. 2013) and myoblast differentiation (Miyamoto et al. 2009). Overall, RBM38 is thought to function as a tumor-suppressor gene in part through its role in stabilizing PTEN and p53 transcripts in renal, liver, and breast cancers and acute myeloid leukemia (Wampfler et al. 2016; Huang et al. 2017; Li et al. 2017c; Wu et al. 2017a; Ye et al. 2018).
In addition to RBM38, the cellular senescence-inhibited gene (CSIG) interacts with the PTEN mRNA in the 5′UTR, impacting PTEN translational regulation (Ma et al. 2008). An interaction between IGF2 mRNA-binding protein 1 (IGF2BP1) and PTEN was shown to be important in directing tumor cell migration through phosphoinositide signaling control (Stöhr et al. 2012). Finally, RBPs including polypyrimidine tract-binding protein 1 (PTBP1), IGF2BP1, cytoplasmic polyadenylation element-binding protein (CPBP), and Staufen1, have been reported to regulate PTEN mRNA stability and translation; however, direct binding data was less than convincing in these studies (Alexandrov et al. 2012; Stöhr et al. 2012; Crawford Parks et al. 2017; Wang et al. 2018). Further work on the relationship of RBPs and PTEN is needed to fully understand their relevance to PTEN biology.
PTEN Regulation by Alternative Splicing and Polyadenylation
Alternative splicing of the PTEN transcript has long been suggested as an important aspect of PTEN regulation and biology (Sharrard and Maitland 2000; Okumura et al. 2011; Oltean and Bates 2014; Malaney et al. 2017). Early studies uncovered two different PTEN splice variants (SVs) in glioblastoma and prostate cancer cells that were termed PTEN-Δ and PTEN-B (Sharrard and Maitland 2000). Both PTEN SVs were demonstrated to generate severe carboxy-terminal truncations in the PTEN protein with little to no phosphatase activity, likely because of decreased stability and rapid degradation. Remarkably, recent work in renal cell carcinoma revealed that PTEN-Δ exerts similar tumor-suppressive effects as full-length PTEN, and that patients with high PTEN-Δ expression had longer metastasis-free and overall survival (Breuksch et al. 2018).
A proportion of individuals with Cowden syndrome (CS) or Bannayan–Riley–Ruvalcaba syndrome (BRRS), the multiple hamartoma conditions associated with germline loss of PTEN, have been found to harbor PTEN SVs, which exclude exons 3, 4, or 6, leading to decreased levels of PTEN protein (Agrawal et al. 2005; Sarquis et al. 2006). These findings were further corroborated by the discovery of PTEN intronic variants in CS and BRRS patients mapping near the splice sites and resulting in exon skipping or activation of cryptic splice sites, eventually resulting in lower expression of PTEN and activation of Akt (Chen et al. 2017). At least eight additional PTEN SVs have been identified, which involve maintaining portions of introns 3 (transcripts 3a, 3b, and 3c) and 5 (transcripts 5a, 5b, and 5c) in the final, processed transcript, as well as SVs that result in the partial deletion of exon 5 (DelE5) or the complete exon 6 (DelE6), and have been found to be differentially expressed in breast cancer (Agrawal and Eng 2006). Other studies have highlighted the inherent complexity of PTEN SVs regulation. For instance, Wang and Chang (1999) found high levels of abnormal PTEN SV transcripts in both normal and tumor-derived tissues, but could not connect them to tumorigenesis (Wang and Chang 1999), whereas Liu et al. (2010) found discrepancies in the detection and proportion of PTEN SVs between fresh and aged mononuclear cells (Liu et al. 2010).
PTEN transcript is also subject to alternative polyadenylation with the main site residing 3 kb downstream of the canonical stop codon (Gray et al. 1998) and the alternative polyadenylation sites 60 and 290 bp downstream of the stop codon, resulting in shorter PTEN transcripts (Hamilton et al. 2000; Xia et al. 2014b). In a study by Thivierge et al., it was hypothesized that shorter PTEN 3′UTRs would limit the access of certain miRNAs and lead to PTEN mRNA stabilization (Thivierge et al. 2018). Surprisingly, this was not observed as longer 3′UTRs were resistant to miRNA-mediated silencing and deemed responsible for the bulk of PTEN protein dosage and signaling functions. Therefore, longer 3′UTRs may possess other features that lead to stabilization and miRNA resistance (Thivierge et al. 2018).
Alternative splicing is a major contributor to transcriptome and proteome diversity; however, deregulated splicing of important cancer genes such as PTEN can lead to a variety of human diseases including cancer. Thus, a better understanding of the mechanisms governing PTEN alternative splicing and the consequences on gene dosage and downstream signaling will shed light on the complex roles of PTEN in tumor suppression and cancer. Importantly, cancer-associated splicing variants of PTEN are potential molecular markers that may be used in prognosis and diagnosis of disease.
PTEN Regulation by mRNA Modification
N6-methyladenosine (m6A) is the most common mRNA modification in eukaryotes (Desrosiers et al. 1974; Wei et al. 1976). An m6A modification entails the covalent addition of a methyl group to an mRNA by a methyltransferase complex comprised of several proteins including METTL3 (Bokar et al. 1997) and METTL14 (Liu et al. 2014). The importance of m6A-modified mRNAs in cancer has only recently been appreciated with the discovery that m6A methylation contributes to tumor suppression and that loss of m6A methylation correlates with malignant, stem-like phenotypes in breast and cervical cancers (Zhang et al. 2016a,b; Wang et al. 2017b). Moreover, overexpression of METTL3 promotes cancer progression and chemoresistance in other cancer types, including hepatic and pancreatic cancers (Chen et al. 2018; Taketo et al. 2018).
To date the role of m6A modification on PTEN regulation is still unclear. However, a single study has shown that METTL3 was able to directly regulate expression of critical downstream effectors, including, but not limited to, c-MYC, BCL-2, and PTEN (Vu et al. 2017). Specifically, loss of METTL3 expression led to reduced m6A methylation of the PTEN transcript and consequently reduced expression of the PTEN protein, suggesting an important role for m6A methylation in the modulation of PTEN expression levels (Vu et al. 2017).
CONCLUDING REMARKS
The variety of mRNA regulatory pathways that converge on PTEN mRNA and impact the levels of PTEN protein are complex and relatively unexplored. Modifications, sequestration, and miRNA-targeting of the PTEN mRNA present an important layer of PTEN regulation in normal and PTEN deregulation in cancer cells (Fig. 2). Importantly, these pathways present possible therapeutic opportunities in attempting to boost PTEN expression in germline carriers of heterozygous mutations, as well as pathologies where PTEN expression is silenced by overactive miRNA silencing. Attempts to inhibit PTEN-targeting miRNAs, but more generally methods to stabilize or restore PTEN mRNA levels (Islam et al. 2018), should be explored as possible therapeutics in cancer, thus adding to the variety of treatment options traditionally focused on interfering with the downstream consequences of PTEN loss.
ACKNOWLEDGMENTS
L.S. is the recipient of a Tier II Canada Research Chair. We thank all members of the Salmena Laboratory for critical discussions.
Footnotes
Editors: Charis Eng, Joanne Ngeow, and Vuk Stambolic
Additional Perspectives on The PTEN Family available at www.perspectivesinmedicine.org
REFERENCES
- Agrawal S, Eng C. 2006. Differential expression of novel naturally occurring splice variants of PTEN and their functional consequences in Cowden syndrome and sporadic breast cancer. Hum Mol Genet 15: 777–787. 10.1093/hmg/ddi492 [DOI] [PubMed] [Google Scholar]
- Agrawal S, Pilarski R, Eng C. 2005. Different splicing defects lead to differential effects downstream of the lipid and protein phosphatase activities of PTEN. Hum Mol Genet 14: 2459–2468. 10.1093/hmg/ddi246 [DOI] [PubMed] [Google Scholar]
- Ala U, Karreth FA, Bosia C, Pagnani A, Taulli R, Léopold V, Tay Y, Provero P, Zecchina R, Pandolfi PP. 2013. Integrated transcriptional and competitive endogenous RNA networks are cross-regulated in permissive molecular environments. Proc Natl Acad Sci 110: 7154–7159. 10.1073/pnas.1222509110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexandrov IM, Ivshina M, Jung DY, Friedline R, Ko HJ, Xu M, O'Sullivan-Murphy B, Bortell R, Huang YT, Urano F, et al. 2012. Cytoplasmic polyadenylation element binding protein deficiency stimulates PTEN and Stat3 mRNA translation and induces hepatic insulin resistance. PLoS Genet 8: e1002457 10.1371/journal.pgen.1002457 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alimonti A, Carracedo A, Clohessy JG, Trotman LC, Nardella C, Egia A, Salmena L, Sampieri K, Haveman WJ, Brogi E, et al. 2010. Subtle variations in Pten dose determine cancer susceptibility. Nat Genet 42: 454–458. 10.1038/ng.556 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartel DP. 2009. MicroRNAs: target recognition and regulatory functions. Cell 136: 215–233. 10.1016/j.cell.2009.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berger AH, Pandolfi PP. 2011. Haplo-insufficiency: a driving force in cancer. J Pathol 223: 137–146. 10.1002/path.2800 [DOI] [PubMed] [Google Scholar]
- Bermúdez Brito M, Goulielmaki E, Papakonstanti EA. 2015. Focus on PTEN regulation. Front Oncol 5: 166 10.3389/fonc.2015.00166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bokar JA, Shambaugh ME, Polayes D, Matera AG, Rottman FM. 1997. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA 3: 1233–1247. [PMC free article] [PubMed] [Google Scholar]
- Bosson AD, Zamudio JR, Sharp PA. 2014. Endogenous miRNA and target concentrations determine susceptibility to potential ceRNA competition. Mol Cell 56: 347–359. 10.1016/j.molcel.2014.09.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breuksch I, Welter J, Bauer H-K, Enklaar T, Frees S, Thüroff JW, Hasenburg A, Prawitt D, Brenner W. 2018. In renal cell carcinoma the PTEN splice variant PTEN-Δ shows similar function as the tumor suppressor PTEN itself. Cell Commun Signal 16: 35 10.1186/s12964-018-0247-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carracedo A, Alimonti A, Pandolfi PP. 2011. PTEN level in tumor suppression: how much is too little? Cancer Res 71: 629–633. 10.1158/0008-5472.CAN-10-2488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A, Bozzoni I. 2011. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 147: 358–369. 10.1016/j.cell.2011.09.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandradoss SD, Schirle NT, Szczepaniak M, MacRae IJ, Joo CA. 2015. A dynamic search process underlies microRNA targeting. Cell 162: 96–107. 10.1016/j.cell.2015.06.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang R-M, Xu J-F, Fang F, Yang H, Yang L-Y. 2016. MicroRNA-130b promotes proliferation and EMT-induced metastasis via PTEN/p-AKT/HIF-1α signaling. Tumour Biol 37: 10609–10619. [DOI] [PubMed] [Google Scholar]
- Chen CL, Tseng YW, Wu JC, Chen GY, Lin KC, Hwang SM, Hu YC. 2015. Suppression of hepatocellular carcinoma by baculovirus-mediated expression of long non-coding RNA PTENP1 and microRNA regulation. Biomaterials 44: 71–81. 10.1016/j.biomaterials.2014.12.023 [DOI] [PubMed] [Google Scholar]
- Chen HJ, Romigh T, Sesock K, Eng C. 2017. Characterization of cryptic splicing in germline PTEN intronic variants in Cowden syndrome. Hum Mutat 38: 1372–1377. 10.1002/humu.23288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen M, Wei L, Law CT, Tsang FHC, Shen J, Cheng CLH, Tsang LH, Ho DWH, Chiu DKC, Lee JMF, et al. 2018. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology 67: 2254–2270. 10.1002/hep.29683 [DOI] [PubMed] [Google Scholar]
- Chi SW, Zang JB, Mele A, Darnell RB. 2009. Argonaute HITS-CLIP decodes microRNA–mRNA interaction maps. Nature 460: 479–486. 10.1038/nature08170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chi H, Yang R, Zheng X, Zhang L, Jiang R, Chen J. 2018. LncRNA RP11-79H23.3 Functions as a competing endogenous RNA to regulate PTEN expression through sponging hsa-miR-107 in the development of bladder cancer. Int J Mol Sci 19: E2531 10.3390/ijms19092531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiu H-S, Martínez MR, Komissarova EV, Llobet-Navas D, Bansal M, Paull EO, Silva J, Yang X, Sumazin P, Califano A. 2018. The number of titrated microRNA species dictates ceRNA regulation. Nucleic Acids Res 46: 4354–4369. 10.1093/nar/gky286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Correia NC, Gírio A, Antunes I, Martins LR, Barata JT. 2014. The multiple layers of non-genetic regulation of PTEN tumour suppressor activity. Eur J Cancer 50: 216–225. 10.1016/j.ejca.2013.08.017 [DOI] [PubMed] [Google Scholar]
- Crawford Parks TE, Ravel-Chapuis A, Bondy-Chorney E, Renaud JM, Côté J, Jasmin BJ. 2017. Muscle-specific expression of the RNA-binding protein Staufen1 induces progressive skeletal muscle atrophy via regulation of phosphatase tensin homolog. Hum Mol Genet 26: 1821–1838. 10.1093/hmg/ddx085 [DOI] [PubMed] [Google Scholar]
- Dahia PL, FitzGerald MG, Zhang X, Marsh DJ, Zheng Z, Pietsch T, von Deimling A, Haluska FG, Haber DA, Eng C. 1998. A highly conserved processed PTEN pseudogene is located on chromosome band 9p21. Oncogene 16: 2403–2406. 10.1038/sj.onc.1201762 [DOI] [PubMed] [Google Scholar]
- Denzler R, Agarwal V, Stefano J, Bartel DP, Stoffel M. 2014. Assessing the ceRNA hypothesis with quantitative measurements of miRNA and target abundance. Mol Cell 54: 766–776. 10.1016/j.molcel.2014.03.045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denzler R, McGeary SE, Title AC, Agarwal V, Bartel DP, Stoffel M. 2016. Impact of microRNA levels, target-site complementarity, and cooperativity on competing endogenous RNA-regulated gene expression. Mol Cell 64: 565–579. 10.1016/j.molcel.2016.09.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desrosiers R, Friderici K, Rottman F. 1974. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci 71: 3971–3975. 10.1073/pnas.71.10.3971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dou L, Meng X, Sui X, Wang S, Shen T, Huang X, Guo J, Fang W, Man Y, Xi J, et al. 2015. MiR-19a regulates PTEN expression to mediate glycogen synthesis in hepatocytes. Sci Rep 5: 11602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang Y, Shen H, Li H, Cao Y, Qin R, Long L, Zhu X, Xie C, Xu W. 2013. miR-106a confers cisplatin resistance by regulating PTEN/Akt pathway in gastric cancer cells. Acta Biochim Biophys Sin (Shanghai) 45: 963–972. [DOI] [PubMed] [Google Scholar]
- Fei D, Zhang X, Liu J, Tan L, Xing J, Zhao D, Zhang Y. 2018. Long noncoding RNA FER1L4 suppresses tumorigenesis by regulating the expression of PTEN targeting miR-18a-5p in osteosarcoma. Cell Physiol Biochem 51: 1364–1375. 10.1159/000495554 [DOI] [PubMed] [Google Scholar]
- Feng YH, Tsao CJ. 2016. Emerging role of microRNA-21 in cancer. Biomed Rep 5: 395–402. 10.3892/br.2016.747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng Y, Zou W, Hu C, Li G, Zhou S, He Y, Ma F, Deng C, Sun L. 2017. Modulation of CASC2/miR-21/PTEN pathway sensitizes cervical cancer to cisplatin. Arch Biochem Biophys 623–624: 20–30. 10.1016/j.abb.2017.05.001 [DOI] [PubMed] [Google Scholar]
- Fujii GH, Morimoto AM, Berson AE, Bolen JB. 1999. Transcriptional analysis of the PTEN/MMAC1 pseudogene, ΨPTEN. Oncogene 18: 1765–1769. 10.1038/sj.onc.1202492 [DOI] [PubMed] [Google Scholar]
- Gao L, Ren W, Zhang L, Li S, Kong X, Zhang H, Dong J, Cai G, Jin C, Zheng D, et al. 2017. PTENp1, a natural sponge of miR-21, mediates PTEN expression to inhibit the proliferation of oral squamous cell carcinoma. Mol Carcinog 56: 1322–1334. 10.1002/mc.22594 [DOI] [PubMed] [Google Scholar]
- Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A, Tacciol C, Pichiorri F, Alder H, Secchiero P, et al. 2009. miR-221 & 222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell 16: 498–509. 10.1016/j.ccr.2009.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Geng L, Sun B, Gao B, Wang Z, Quan C, Wei F, Fang X-D. 2014. MicroRNA-103 promotes colorectal cancer by targeting tumor suppressor DICER and PTEN. Int J Mol Sci 15: 8458–8472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong L, Zhang W, Yuan Y, Xing X, Li H, Zhao G. 2018. miR-222 promotes invasion and migration of ovarian carcinoma by targeting PTEN. Oncol Lett 16: 984–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gray IC, Stewart LM, Phillips SM, Hamilton JA, Gray NE, Watson GJ, Spurr NK, Snary D. 1998. Mutation and expression analysis of the putative prostate tumour-suppressor gene PTEN. Br J Cancer 78: 1296–1300. 10.1038/bjc.1998.674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grimson A, Farh KKH, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP. 2007. MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 27: 91–105. 10.1016/j.molcel.2007.06.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo X, Deng L, Deng K, Wang H, Shan T, Zhou H, Liang Z, Xia J, Li C. 2016. Pseudogene PTENP1 suppresses gastric cancer progression by modulating PTEN. Anticancer Agents Med Chem 16: 456–464. 10.2174/1871520615666150507121407 [DOI] [PubMed] [Google Scholar]
- Hafner M, Landthaler M, Burger L, Khorshid M, Hausser J, Berninger P, Rothballer A, Ascano M Jr, Jungkamp AC, Munschauer M, et al. 2010. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 141: 129–141. 10.1016/j.cell.2010.03.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamilton JA, Stewart LM, Ajayi L, Gray IC, Gray NE, Roberts KG, Watson GJ, Kaisary AV, Snary D. 2000. The expression profile for the tumour suppressor gene PTEN and associated polymorphic markers. Br J Cancer 82: 1671–1676. 10.1054/bjoc.2000.1211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- He Y, Bai J, Liu P, Dong J, Tang Y, Zhou J, Han P, Xing J, Chen Y, Yu X. 2017. miR-494 protects pancreatic β-cell function by targeting PTEN in gestational diabetes mellitus. EXCLI J 16: 1297–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heinicke LA, Nabet B, Shen S, Jiang P, van Zalen S, Cieply B, Russell JE, Xing Y, Carstens RP. 2013. The RNA binding protein RBM38 (RNPC1) regulates splicing during late erythroid differentiation. PLoS ONE 8: e78031 10.1371/journal.pone.0078031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hopkins BD, Fine B, Steinbach N, Dendy M, Rapp Z, Shaw J, Pappas K, Yu JS, Hodakoski C, Mense S, et al. 2013. A secreted PTEN phosphatase that enters cells to alter signaling and survival. Science 341: 399–402. 10.1126/science.1234907 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu YL, Hung JY, Chang WA, Jian SF, Lin YS, Pan YC, Wu CY, Kuo PL. 2018. Hypoxic lung-cancer-derived extracellular vesicle microRNA-103a increases the oncogenic effects of macrophages by targeting PTEN. Mol Ther 26: 568–581. 10.1016/j.ymthe.2017.11.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang W, Wei XL, Ni W, Cao M, Meng L, Yang H. 2017. The expression of RNA-binding protein RBM38 decreased in renal cell carcinoma and represses renal cancer cell proliferation, migration, and invasion. Tumour Biol 39: 1010428317701635. [DOI] [PubMed] [Google Scholar]
- Huse JT, Brennan C, Hambardzumyan D, Wee B, Pena J, Rouhanifard SH, Sohn-Lee C, le Sage C, Agami R, Tuschl T, et al. 2009. The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo. Genes Dev 23: 1327–1337. 10.1101/gad.1777409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ioffe YJ, Chiappinelli KB, Mutch DG, Zighelboim I, Goodfellow PJ. 2012. Phosphatase and tensin homolog (PTEN) pseudogene expression in endometrial cancer: a conserved regulatory mechanism important in tumorigenesis? Gynecol Oncol 124: 340–346. 10.1016/j.ygyno.2011.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Islam MA, Xu Y, Tao W, Ubellacker JM, Lim M, Aum D, Lee GY, Zhou K, Zope H, Yu M, et al. 2018. Restoration of tumor-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat Biomed Eng 2: 850–864. 10.1038/s41551-018-0284-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ji R, Cheng Y, Yue J, Yang J, Liu X, Chen H, Dean DB, Zhang C. 2007. MicroRNA expression signature and antisense-mediated depletion reveal an essential role of microRNA in vascular neointimal lesion formation. Circ Res 100: 1579–1588. 10.1161/CIRCRESAHA.106.141986 [DOI] [PubMed] [Google Scholar]
- Jia Z, Peng J, Yang Z, Chen J, Liu L, Luo D, He P. 2019. Long non-coding RNA TP73-AS1 promotes colorectal cancer proliferation by acting as a ceRNA for miR-103 to regulate PTEN expression. Gene 685: 222–229. 10.1016/j.gene.2018.11.072 [DOI] [PubMed] [Google Scholar]
- Jiang Y, Chang H, Chen G. 2018. Effects of microRNA-20a on the proliferation, migration and apoptosis of multiple myeloma via the PTEN/PI3K/AKT signaling pathway. Oncol Lett 15: 10001–10007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jonas S, Izaurralde E. 2015. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet 16: 421–433. 10.1038/nrg3965 [DOI] [PubMed] [Google Scholar]
- Kakizaki T, Hatakeyama H, Nakamaru Y, Takagi D, Mizumachi T, Sakashita T, Kano S, Homma A, Fukuda S. 2017. Role of microRNA-296-3p in the malignant transformation of sinonasal inverted papilloma. Oncol Lett 14: 987–992. 10.3892/ol.2017.6193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karreth FA, Tay Y, Perna D, Ala U, Tan SM, Rust AG, DeNicola G, Webster KA, Weiss D, Perez-Mancera PA, et al. 2011. In vivo identification of tumor-suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma. Cell 147: 382–395. 10.1016/j.cell.2011.09.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kato M, Putta S, Wang M, Yuan H, Lanting L, Nair I, Gunn A, Nakagawa Y, Shimano H, Todorov I, et al. 2009. TGF-β activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN. Nat Cell Biol 11: 881–889. 10.1038/ncb1897 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kehl T, Backes C, Kern F, Fehlmann T, Ludwig N, Meese E, Lenhof H-P, Keller A. 2017. About miRNAs, miRNA seeds, target genes and target pathways. Oncotarget 8: 107167–107175. 10.18632/oncotarget.22363 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovalenko TF, Sorokina AV, Ozolinia LA, Patrushev LI. 2013. Pseudogene PTENP1 5′-region methylation in endometrial cancer and hyperplasias. Bioorg Khim 39: 445–453. [DOI] [PubMed] [Google Scholar]
- Kovalenko TF, Morozova KV, Ozolinya LA, Lapina IA, Patrushev LI. 2018. The PTENP1 pseudogene, unlike the PTEN gene, is methylated in normal endometrium, as well as in endometrial hyperplasias and carcinomas in middle-aged and elderly females. Acta Naturae 10: 43–50. 10.32607/20758251-2018-10-1-43-50 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwabi-Addo B, Giri D, Schmidt K, Podsypanina K, Parsons R, Greenberg N, Ittmann M. 2001. Haploinsufficiency of the Pten tumor suppressor gene promotes prostate cancer progression. Proc Natl Acad Sci 98: 11563–11568. 10.1073/pnas.201167798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai Y, He S, Ma L, Lin H, Ren B, Ma J, Zhu X, Zhuang S. 2017. HOTAIR functions as a competing endogenous RNA to regulate PTEN expression by inhibiting miR-19 in cardiac hypertrophy. Mol Cell Biochem 432: 179–187. 10.1007/s11010-017-3008-y [DOI] [PubMed] [Google Scholar]
- Lee DY, Jeyapalan Z, Fang L, Yang J, Zhang Y, Yee AY, Li M, Du WW, Shatseva T, Yang BB. 2010. Expression of versican 3′-untranslated region modulates endogenous microRNA functions. PLoS ONE 5: e13599 10.1371/journal.pone.0013599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee YR, Chen M, Pandolfi PP. 2018. The functions and regulation of the PTEN tumor suppressor: new modes and prospects. Nat Rev Mol Cell Biol 19: 547–562. 10.1038/s41580-018-0015-0 [DOI] [PubMed] [Google Scholar]
- Léveillé N, Elkon R, Davalos V, Manoharan V, Hollingworth D, Vrielink JO, le Sage C, Melo CA, Horlings HM, Wesseling J, et al. 2011. Selective inhibition of microRNA accessibility by RBM38 is required for p53 activity. Nat Commun 2: 513 10.1038/ncomms1519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li N, Miao Y, Shan Y, Liu B, Li Y, Zhao L, Jia L. 2017a. MiR-106b and miR-93 regulate cell progression by suppression of PTEN via PI3 K/Akt pathway in breast cancer. Cell Death Dis 8: e2796 10.1038/cddis.2017.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li RK, Gao J, Guo LH, Huang GQ, Luo WH. 2017b. PTENP1 acts as a ceRNA to regulate PTEN by sponging miR-19b and explores the biological role of PTENP1 in breast cancer. Cancer Gene Ther 24: 309–315. 10.1038/cgt.2017.29 [DOI] [PubMed] [Google Scholar]
- Li XX, Shi L, Zhou XJ, Wu J, Xia TS, Zhou WB, Sun X, Zhu L, Wei JF, Ding Q. 2017c. The role of c-Myc-RBM38 loop in the growth suppression in breast cancer. J Exp Clin Cancer Res 36: 49 10.1186/s13046-017-0521-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Yan X, Wang F, Yang Q, Luo X, Kong J, Ju S. 2019. Down-regulated lncRNA SLC25A5-AS1 facilitates cell growth and inhibits apoptosis via miR-19a-3p/PTEN/PI3K/AKT signalling pathway in gastric cancer. J Cell Mol Med 23: 2920–2932. 10.1111/jcmm.14200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin H, Huang ZP, Liu J, Qiu Y, Tao YP, Wang MC, Yao H, Hou KZ, Gu FM, Xu XF. 2018. MiR-494-3p promotes PI3 K/AKT pathway hyperactivation and human hepatocellular carcinoma progression by targeting PTEN. Sci Rep 8: 10461 10.1038/s41598-018-28519-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ling S, Birnbaum Y, Nanhwan MK, Thomas B, Bajaj M, Li Y, Li Y, Ye Y. 2013. Dickkopf-1 (DKK1) phosphatase and tensin homolog on chromosome 10 (PTEN) crosstalk via microRNA interference in the diabetic heart. Basic Res Cardiol 108: 352 10.1007/s00395-013-0352-2 [DOI] [PubMed] [Google Scholar]
- Liu Y, Malaviarachchi P, Beggs M, Emanuel PD. 2010. PTEN transcript variants caused by illegitimate splicing in “aged” blood samples and EBV-transformed cell lines. Hum Genet 128: 609–614. 10.1007/s00439-010-0886-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C, Mallick B, Long D, Rennie WA, Wolenc A, Carmack CS, Ding Y. 2013a. CLIP-based prediction of mammalian microRNA binding sites. Nucleic Acids Res 41: e138 10.1093/nar/gkt435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu ZL, Wang H, Liu J, Wang ZX. 2013b. MicroRNA-21 (miR-21) expression promotes growth, metastasis, and chemo- or radioresistance in non-small cell lung cancer cells by targeting PTEN. Mol Cell Biochem 372: 35–45. 10.1007/s11010-012-1443-3 [DOI] [PubMed] [Google Scholar]
- Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al. 2014. A METTL3–METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 10: 93–95. 10.1038/nchembio.1432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu L, Li Y, Liu S, Duan Q, Chen L, Wu T, Qian H, Yang S, Xin D. 2017. Downregulation of miR-193a-3p inhibits cell growth and migration in renal cell carcinoma by targeting PTEN. Tumour Biol 39: 1010428317711951. [DOI] [PubMed] [Google Scholar]
- Lozano-Bartolomé J, Llauradó G, Portero-Otin M, Altuna-Coy A, Rojo-Martínez G, Vendrell J, Jorba R, Rodríguez-Gallego E, Chacón MR. 2018. Altered expression of miR-181a-5p and miR-23a-3p is associated with obesity and TNFα-induced insulin resistance. J Clin Endocrinol Metab 103: 1447–1458. 10.1210/jc.2017-01909 [DOI] [PubMed] [Google Scholar]
- Lu E, Su J, Zeng W, Zhang C. 2016. Enhanced miR-9 promotes laryngocarcinoma cell survival via down-regulating PTEN. Biomed Pharmacother 84: 608–613. 10.1016/j.biopha.2016.09.047 [DOI] [PubMed] [Google Scholar]
- Lu J, Mu X, Yin Q, Hu K. 2019. miR-106a contributes to prostate carcinoma progression through PTEN. Oncol Lett 17: 1327–1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma L, Chang N, Guo S, Li Q, Zhang Z, Wang W, Tong T. 2008. CSIG inhibits PTEN translation in replicative senescence. Mol Cell Biol 2008: 6290–301. 10.1128/MCB.00142-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malaney P, Uversky VN, Davé V. 2017. PTEN proteoforms in biology and disease. Cell Mol Life Sci 74: 2783–2794. 10.1007/s00018-017-2500-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marsit CJ, Zheng S, Aldape K, Hinds PW, Nelson HH, Wiencke JK, Kelsey KT. 2005. PTEN expression in non-small-cell lung cancer: evaluating its relation to tumor characteristics, allelic loss, and epigenetic alteration. Hum Pathol 36: 768–776. 10.1016/j.humpath.2005.05.006 [DOI] [PubMed] [Google Scholar]
- Meng F, Henson R, Lang M, Wehbe H, Maheshwari S, Mendell JT, Jiang J, Schmittgen TD, Patel T. 2006. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology 130: 2113–2129. 10.1053/j.gastro.2006.02.057 [DOI] [PubMed] [Google Scholar]
- Meng F, Henson R, Wehbe–Janek H, Ghoshal K, Jacob ST, Patel T. 2007. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 133: 647–658. 10.1053/j.gastro.2007.05.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miao Y, Zheng W, Li N, Su Z, Zhao L, Zhou H, Jia L. 2017. MicroRNA-130b targets PTEN to mediate drug resistance and proliferation of breast cancer cells via the PI3 K/Akt signaling pathway. Sci Rep 7: 41942 10.1038/srep41942 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyamoto S, Hidaka K, Jin D, Morisaki T. 2009. RNA-binding proteins Rbm38 and Rbm24 regulate myogenic differentiation via p21-dependent and -independent regulatory pathways. Genes Cells 14: 1241–1252. 10.1111/j.1365-2443.2009.01347.x [DOI] [PubMed] [Google Scholar]
- Nie X, Fan J, Li H, Yin Z, Zhao Y, Dai B, Dong N, Chen C, Wang DW. 2018. miR-217 promotes cardiac hypertrophy and dysfunction by targeting PTEN. Mol Ther Nucleic Acids 12: 254–266. 10.1016/j.omtn.2018.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okumura N, Yoshida H, Kitagishi Y, Nishimura Y, Matsuda S. 2011. Alternative splicings on p53, BRCA1 and PTEN genes involved in breast cancer. Biochem Biophys Res Commun 413: 395–399. 10.1016/j.bbrc.2011.08.098 [DOI] [PubMed] [Google Scholar]
- Oltean S, Bates DO. 2014. Hallmarks of alternative splicing in cancer. Oncogene 33: 5311–5318. 10.1038/onc.2013.533 [DOI] [PubMed] [Google Scholar]
- Peterson SM, Thompson JA, Ufkin ML, Sathyanarayana P, Liaw L, Congdon CB. 2014. Common features of microRNA target prediction tools. Front Genet 5: 23 10.3389/fgene.2014.00023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poliseno L, Salmena L, Riccardi L, Fornari A, Song MS, Hobbs RM, Sportoletti P, Varmeh S, Egia A, Fedele G, et al. 2010a. Identification of the miR-106b∼25 microRNA cluster as a proto-oncogenic PTEN-targeting intron that cooperates with its host gene MCM7 in transformation. Sci Signal 3: ra29 10.1126/scisignal.2000594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. 2010b. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465: 1033–1038. 10.1038/nature09144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poliseno L, Haimovic A, Christos PJ, Vega Y, Vega y Saenz de Miera EC, Shapiro R, Pavlick A, Berman RS, Darvishian F, Osman I. 2011. Deletion of PTENP1 pseudogene in human melanoma. J Invest Dermatol 131: 2497–2500. 10.1038/jid.2011.232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollutri D, Patrizi C, Marinelli S, Giovannini C, Trombetta E, Giannone FA, Baldassarre M, Quarta S, Vandewynckel YP, Vandierendonck A, et al. 2018. The epigenetically regulated miR-494 associates with stem-cell phenotype and induces sorafenib resistance in hepatocellular carcinoma. Cell Death Dis 9: 4 10.1038/s41419-017-0076-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qian YY, Li K, Liu QY, Liu ZS. 2017. Long non-coding RNA PTENP1 interacts with miR-193a-3p to suppress cell migration and invasion through the PTEN pathway in hepatocellular carcinoma. Oncotarget 8: 107859–107869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin N, Tong GF, Sun LW, Xu XL. 2017. Long noncoding RNA MEG3 suppresses glioma cell proliferation, migration, and invasion by acting as a competing endogenous RNA of miR-19a. Oncol Res 25: 1471–1478. 10.3727/096504017X14886689179993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin Y, Huo Z, Song X, Chen X, Tian X, Wang X. 2018. mir-106a regulates cell proliferation and apoptosis of colon cancer cells through targeting the PTEN/PI3 K/AKT signaling pathway. Oncol Lett 15: 3197–3201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren P, Gong F, Zhang Y, Jiang J, Zhang H. 2016. MicroRNA-92a promotes growth, metastasis, and chemoresistance in non-small cell lung cancer cells by targeting PTEN. Tumour Biol 37: 3215–3225. 10.1007/s13277-015-4150-3 [DOI] [PubMed] [Google Scholar]
- Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. 2011. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell 146: 353–358. 10.1016/j.cell.2011.07.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarquis MS, Agrawal S, Shen L, Pilarski R, Zhou XP, Eng C. 2006. Distinct expression profiles for PTEN transcript and its splice variants in Cowden syndrome and Bannayan–Riley–Ruvalcaba syndrome. Am J Hum Genet 79: 23–30. 10.1086/504392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seitz H. 2009. Redefining microRNA targets. Curr Biol 19: 870–873. 10.1016/j.cub.2009.03.059 [DOI] [PubMed] [Google Scholar]
- Sharrard RM, Maitland NJ. 2000. Alternative splicing of the human PTEN/MMAC1/TEP1 gene. Biochim Biophys Acta 1494: 282–285. 10.1016/S0167-4781(00)00210-4 [DOI] [PubMed] [Google Scholar]
- Shi X, Tang X, Su L. 2018. Overexpression of long noncoding RNA PTENP1 inhibits cell proliferation and migration via suppression of miR-19b in breast cancer cells. Oncol Res 26: 869–878. 10.3727/096504017X15123838050075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song MS, Salmena L, Pandolfi PP. 2012. The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol 13: 283–296. 10.1038/nrm3330 [DOI] [PubMed] [Google Scholar]
- Stöhr N, Köhn M, Lederer M, Glass M, Reinke C, Singer RH, Hüttelmaier S. 2012. IGF2BP1 promotes cell migration by regulating MK5 and PTEN signaling. Genes Dev 26: 176–189. 10.1101/gad.177642.111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sui J, Yang X, Qi W, Guo K, Gao Z, Wang L, Sun D. 2017. Long non-coding RNA Linc-USP16 functions as a tumour suppressor in hepatocellular carcinoma by regulating PTEN expression. Cell Physiol Biochem 44: 1188–1198. 10.1159/000485449 [DOI] [PubMed] [Google Scholar]
- Sumazin P, Yang X, Chiu H-S, Chung W-J, Iyer A, Llobet-Navas D, Rajbhandari P, Bansal M, Guarnieri P, Silva J, et al. 2011. An extensive microRNA-mediated network of RNA-RNA interactions regulates established oncogenic pathways in glioblastoma. Cell 147: 370–381. 10.1016/j.cell.2011.09.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taketo K, Konno M, Asai A, Koseki J, Toratani M, Satoh T, Doki Y, Mori M, Ishii H, Ogawa K. 2018. The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol 52: 621–629. [DOI] [PubMed] [Google Scholar]
- Tay Y, Kats L, Salmena L, Weiss D, Tan SM, Ala U, Karreth F, Poliseno L, Provero P, Di Cunto F, et al. 2011. Coding-independent regulation of the tumor suppressor PTEN by competing endogenous mRNAs. Cell 147: 344–357. 10.1016/j.cell.2011.09.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thivierge C, Tseng HW, Mayya VK, Lussier C, Gravel SP, Duchaine TF. 2018. Alternative polyadenylation confers Pten mRNAs stability and resistance to microRNAs. Nucleic Acids Res 46: 10340–10352. 10.1093/nar/gky666 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trotman LC, Niki M, Dotan ZA, Koutcher JA, Di Cristofano A, Xiao A, Khoo AS, Roy-Burman P, Greenberg NM, Van Dyke T, et al. 2003. Pten dose dictates cancer progression in the prostate. PLoS Biol 1: E59 10.1371/journal.pbio.0000059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vu LP, Pickering BF, Cheng Y, Zaccara S, Nguyen D, Minuesa G, Chou T, Chow A, Saletore Y, MacKay M, et al. 2017. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 23: 1369–1376. 10.1038/nm.4416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wampfler J, Federzoni EA, Torbett BE, Fey MF, Tschan MP. 2016. The RNA binding proteins RBM38 and DND1 are repressed in AML and have a novel function in APL differentiation. Leuk Res 41: 96–102. 10.1016/j.leukres.2015.12.006 [DOI] [PubMed] [Google Scholar]
- Wang NM, Chang JG. 1999. Are aberrant transcripts of FHIT, TSG101, and PTEN/MMAC1 oncogenesis related? Int J Mol Med 3: 491–495. [DOI] [PubMed] [Google Scholar]
- Wang C, Huai L, Zhang C, Jia Y, Li Q, Chen Y, Tian Z, Tang K, Xing H, Wang M, et al. 2012. Study on expression of PTEN gene and its pseudogene PTENP1 in acute leukemia and correlation between them. Zhonghua Xue Ye Xue Za Zhi 33: 896–901. [PubMed] [Google Scholar]
- Wang D, Wang Y, Ma J, Wang W, Sun B, Zheng T, Wei M, Sun Y. 2017a. MicroRNA-20a participates in the aerobic exercise-based prevention of coronary artery disease by targeting PTEN. Biomed Pharmacother 95: 756–763. 10.1016/j.biopha.2017.08.086 [DOI] [PubMed] [Google Scholar]
- Wang X, Li Z, Kong B, Song C, Cong J, Hou J, Wang S. 2017b. Reduced m6A mRNA methylation is correlated with the progression of human cervical cancer. Oncotarget 8: 98918–98930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Li Y, Fan Y, Yu X, Mao X, Jin F. 2018. PTBP1 promotes the growth of breast cancer cells through the PTEN/Akt pathway and autophagy. J Cell Physiol 233: 8930–8939. 10.1002/jcp.26823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei CM, Gershowitz A, Moss B. 1976. 5′-Terminal and internal methylated nucleotide sequences in HeLa cell mRNA. Biochemistry 15: 397–401. 10.1021/bi00647a024 [DOI] [PubMed] [Google Scholar]
- Whang YE, Wu X, Sawyers CL. 1998. Identification of a pseudogene that can masquerade as a mutant allele of the PTEN/MMAC1 tumor suppressor gene. J Natl Cancer Inst 90: 859–861. 10.1093/jnci/90.11.859 [DOI] [PubMed] [Google Scholar]
- Wu J, Zhou XJ, Sun X, Xia TS, Li XX, Shi L, Zhu L, Zhou WB, Wei JF, Ding Q. 2017a. RBM38 is involved in TGF-β-induced epithelial-to-mesenchymal transition by stabilising zonula occludens-1 mRNA in breast cancer. Br J Cancer 117: 675–684. 10.1038/bjc.2017.204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia H, Ooi LLPJ, Hui KM. 2013. MicroRNA-216a/217-induced epithelial-mesenchymal transition targets PTEN and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology 58: 629–641. 10.1002/hep.26369 [DOI] [PubMed] [Google Scholar]
- Xia T, Liao Q, Jiang X, Shao Y, Xiao B, Xi Y, Guo J. 2014a. Long noncoding RNA associated-competing endogenous RNAs in gastric cancer. Sci Rep 4: 6088 10.1038/srep06088 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia Z, Donehower LA, Cooper TA, Neilson JR, Wheeler DA, Wagner EJ, Li W. 2014b. Dynamic analyses of alternative polyadenylation from RNA-seq reveal a 3′-UTR landscape across seven tumour types. Nat Commun 5: 5274 10.1038/ncomms6274 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia T, Chen S, Jiang Z, Shao Y, Jiang X, Li P, Xiao B, Guo J. 2015. Long noncoding RNA FER1L4 suppresses cancer cell growth by acting as a competing endogenous RNA and regulating PTEN expression. Sci Rep 5: 13445 10.1038/srep13445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao C, Srinivasan L, Calado DP, Patterson HC, Zhang B, Wang J, Henderson JM, Kutok JL, Rajewsky K. 2008. Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat Immunol 9: 405–414. 10.1038/ni1575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao J, Yu W, Hu K, Li M, Chen J, Li Z. 2017. miR-92a promotes tumor growth of osteosarcoma by targeting PTEN/AKT signaling pathway. Oncol Rep 37: 2513–2521. 10.3892/or.2017.5484 [DOI] [PubMed] [Google Scholar]
- Xie Q, Lin S, Zheng M, Cai Q, Tu Y. 2019. Long noncoding RNA NEAT1 promotes the growth of cervical cancer cells via sponging miR-9-5p. Biochem Cell Biol 97: 100–108. 10.1139/bcb-2018-0111 [DOI] [PubMed] [Google Scholar]
- Xu J, Tang Y, Bei Y, Ding S, Che L, Yao J, Wang H, Lv D, Xiao J. 2016. miR-19b attenuates H2O2-induced apoptosis in rat H9C2 cardiomyocytes via targeting PTEN. Oncotarget 7: 10870–10878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H, Kong W, He L, Zhao J-J, O'Donnell JD, Wang J, Wenham RM, Coppola D, Kruk PA, Nicosia SV, et al. 2008. MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res 68: 425–433. 10.1158/0008-5472.CAN-07-2488 [DOI] [PubMed] [Google Scholar]
- Yang H, Zheng W, Zhao W, Guan C, An J. 2013. Roles of miR-590-5p and miR-590-3p in the development of hepatocellular carcinoma. Nan Fang Yi Ke Da Xue Xue Bao 33: 804–811. [PubMed] [Google Scholar]
- Yang L, Li Y, Gong R, Gao M, Feng C, Liu T, Sun Y, Jin M, Wang D, Yuan Y, et al. 2018. The long noncoding RNA-ORLNC1 regulates bone mass by directing mesenchymal stem cell fate. Mol Ther 27: 394–410. 10.1016/j.ymthe.2018.11.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye J, Liang R, Bai T, Lin Y, Mai R, Wei M, Ye X, Li L, Wu F. 2018. RBM38 plays a tumor-suppressor role via stabilizing the p53-mdm2 loop function in hepatocellular carcinoma. J Exp Clin Cancer Res 37: 212 10.1186/s13046-018-0852-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yndestad S, Austreid E, Knappskog S, Chrisanthar R, Lilleng PK, Lønning PE, Eikesdal HP. 2017. High PTEN gene expression is a negative prognostic marker in human primary breast cancers with preserved p53 function. Breast Cancer Res Treat 163: 177–190. 10.1007/s10549-017-4160-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yndestad S, Austreid E, Skaftnesmo KO, Lønning PE, Eikesdal HP. 2018. Divergent activity of the pseudogene PTENP1 in ER-positive and negative breast cancer. Mol Cancer Res 16: 78–89. 10.1158/1541-7786.MCR-17-0207 [DOI] [PubMed] [Google Scholar]
- Yu G, Yao W, Gumireddy K, Li A, Wang J, Xiao W, Chen K, Xiao H, Li H, Tang K, et al. 2014. Pseudogene PTENP1 functions as a competing endogenous RNA to suppress clear-cell renal cell carcinoma progression. Mol Cancer Ther 13: 3086–3097. 10.1158/1535-7163.MCT-14-0245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu G, Ou ZY, Tao QY, Wan GY, Lu ZH, Lang B. 2017. Role of lncRNA PTENP1 in tumorigenesis and progression of bladder cancer and the molecular mechanism. Nan Fang Yi Ke Da Xue Xue Bao 37: 1494–1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu W, Li D, Ding X, Sun Y, Liu Y, Cong J, Yang J, Sun J, Ning X, Wang H, et al. 2019. LINC00702 suppresses proliferation and invasion in small non-cell lung cancer through regulating miR-510/PTEN axis. Aging (Albany NY) 11: 1471–1485. 10.18632/aging.101846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yue B, Sun B, Liu C, Zhao S, Zhang D, Yu F, Yan D. 2015. Long non-coding RNA Fer-1-like protein 4 suppresses oncogenesis and exhibits prognostic value by associating with miR-106a-5p in colon cancer. Cancer Sci 106: 1323–1332. 10.1111/cas.12759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zaman MS, Thamminana S, Shahryari V, Chiyomaru T, Deng G, Saini S, Majid S, Fukuhara S, Chang I, Arora S, et al. 2012. Inhibition of PTEN gene expression by oncogenic miR-23b-3p in renal cancer. PLoS ONE 7: e50203 10.1371/journal.pone.0050203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zarringhalam K, Tay Y, Kulkarni P, Bester AC, Pandolfi PP, Kulkarni RV. 2017. Identification of competing endogenous RNAs of the tumor suppressor gene PTEN: a probabilistic approach. Sci Rep 7: 7755 10.1038/s41598-017-08209-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Wang J, Zhao F, Liu Q, Jiang K, Yang G. 2010. MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin Chim Acta 411: 846–852. 10.1016/j.cca.2010.02.074 [DOI] [PubMed] [Google Scholar]
- Zhang Z, Zhu Z, Watabe K, Zhang X, Bai C, Xu M, Wu F, Mo YY. 2013. Negative regulation of lncRNA GAS5 by miR-21. Cell Death Differ 20: 1558–1568. 10.1038/cdd.2013.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Zheng L, Ding Y, Li Q, Wang R, Liu T, Sun Q, Yang H, Peng S, Wang W, et al. 2015. MiR-20a induces cell radioresistance by activating the PTEN/PI3 K/Akt signaling pathway in hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 92: 1132–1140. 10.1016/j.ijrobp.2015.04.007 [DOI] [PubMed] [Google Scholar]
- Zhang C, Samanta D, Lu H, Bullen JW, Zhang H, Chen I, He X, Semenza GL. 2016a. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc Natl Acad Sci 113: E2047–E2056. 10.1073/pnas.1602883113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C, Zhi WI, Lu H, Samanta D, Chen I, Gabrielson E, Semenza GL. 2016b. Hypoxia-inducible factors regulate pluripotency factor expression by ZNF217- and ALKBH5-mediated modulation of RNA methylation in breast cancer cells. Oncotarget 7: 64527–64542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang R, Guo Y, Ma Z, Ma G, Xue Q, Li F, Liu L. 2017. Long non-coding RNA PTENP1 functions as a ceRNA to modulate PTEN level by decoying miR-106b and miR-93 in gastric cancer. Oncotarget 8: 26079–26089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Xu E, Ren C, Yang HJ, Zhang Y, Sun W, Kong X, Zhang W, Chen M, Huang E, et al. 2018a. Genetic ablation of Rbm38 promotes lymphomagenesis in the context of mutant p53 by downregulating PTEN. Cancer Res 78: 1511–1521. 10.1158/0008-5472.CAN-17-2457 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang XF, Ye Y, Zhao S-J. 2018b. LncRNA Gas5 acts as a ceRNA to regulate PTEN expression by sponging miR-222-3p in papillary thyroid carcinoma. Oncotarget 9: 3519–3530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang B, Liu Y, Zhang J. 2019. Silencing of miR-19a-3p enhances osteosarcoma cells chemosensitivity by elevating the expression of tumor suppressor PTEN. Oncol Lett 17: 414–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao D, Chen Y, Chen S, Zheng C, Hu J, Luo S. 2017. MiR-19a regulates the cell growth and apoptosis of osteosarcoma stem cells by targeting PTEN. Tumour Biol 39: 1010428317705341. [DOI] [PubMed] [Google Scholar]
- Zhou S, Shen D, Wang Y, Gong L, Tang X, Yu B, Gu X, Ding F. 2012. MicroRNA-222 targeting PTEN promotes neurite outgrowth from adult dorsal root ganglion neurons following sciatic nerve transection. PLoS ONE 7: e44768 10.1371/journal.pone.0044768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou XJ, Wu J, Shi L, Li XX, Zhu L, Sun X, Qian JY, Wang Y, Wei JF, Ding Q. 2017. PTEN expression is upregulated by an RNA-binding protein RBM38 via enhancing its mRNA stability in breast cancer. J Exp Clin Cancer Res 36: 149 10.1186/s13046-017-0620-3 [DOI] [PMC free article] [PubMed] [Google Scholar]