Abstract
The mechanistic target of rapamycin (mTOR) is a major signaling hub that coordinates cellular and organismal responses, such as cell growth, proliferation, apoptosis, and metabolism. Dysregulation of mTOR signaling occurs in many human diseases, and there are significant ongoing efforts to pharmacologically target this pathway. Long noncoding RNAs (lncRNA), defined by a length > 200 nucleotides and absence of a long open-reading-frame, are a class of non-protein-coding RNAs. Mutations and dysregulations of lncRNAs are directly linked to the development and progression of many diseases, including cancer, diabetes, and neurologic disorders. Recent findings reveal diverse functions for lncRNA that include transcriptional regulation, organization of nuclear domains, and regulation of proteins or RNA molecules. Despite considerable development in our understanding of lncRNA over the past decade, only a fraction of annotated lncRNAs has been examined for biological function. In addition, lncRNAs have emerged as therapeutic targets due to their ability to modulate multiple pathways, including mTOR signaling. This review will provide an up-to-date summary of lncRNAs that are involved in regulating mTOR pathway.
1. Introduction
The mTOR pathway is an evolutionary conserved signaling pathway that senses and integrates extra- and intra-cellular signals to coordinate basic cellular and organismal responses such as cell growth, proliferation, apoptosis, and metabolism [1]. mTOR is one of the most frequently dysregulated signaling cascades in human diseases and malignancies [2,3]. mTOR is a serine/threonine protein kinase in the PI3K-related family that forms the catalytic subunit of two structurally and functionally distinct protein complexes known as mTOR Complex 1 (mTORC1) and mTOR Complex 2 (mTORC2) [4,5]. mTOR is activated by diverse stimuli such as growth factors [6], nutrients [7], and energy and stress signals [8] in order to control cell growth, proliferation, and survival. mTORC1 has five components: mTOR, mLST8, Raptor, PRAS40, and Deptor (Fig. 1A), and is sensitive to the immunosuppressant and anticancer drug rapamycin. mTORC1 controls gene transcription, ribosomal biogenesis, protein synthesis, autophagy, and other growth-related processes [9]. Downstream bona fide targets of mTORC1 include S6 kinase 1 (S6K1) and eIF4E-binding protein 1 (4E-BP1) [10]. mTORC2 has six components: mTOR, Rictor, mSIN1, Protor, mLST8, and Deptor (Fig. 1B). The function of mTORC2 is less well understood and is thought to promote cell survival and organization of the actin cytoskeleton [11]. mTORC2 is resistant to rapamycin and generally insensitive to nutrients and energy signals. Cell survival is mediated primarily through activation of AKT phosphorylation at Ser473 [12], as well as other members of the protein kinases A, G, and C family [13]. Activation of mTORC1 occurs by upstream regulators, such as phosphatidylinositol 3-kinase (PI3K), PDK1, TSC1-TSC2 complex, and Rheb [14]. However, the mechanism by which growth factors activate mTORC2 is not clear. The crucial role of mTOR in cell biology has generated interest in mTOR inhibitors, many of which have already undergone clinical trials for disease treatment, mainly cancer. Limitations of mTOR inhibitors, such as rapamycin and many of its analogs, include toxicity, sensitivity, and unwanted side effects such as hyperinsulinemia, glucose intolerance, and dysregulated autophagy [15]. Limitations also arise at the molecular level; for example, hyperphosphorylation of 4E-BP1, which is pathogenic in human cancer, is usually resistant to rapamycin/rapalogs [16]. Many proteins and signaling molecules are known to control mTOR activity; however, recent studies have identified long non-coding RNAs (lncRNAs) as novel regulators of the mTOR pathway.
Fig. 1.
The mTOR signaling pathway. Protein components and downstream effectors of A) mTORC1 and B) mTORC2.
Although a large number of RNA species are transcribed from the mammalian genome, only a small fraction of the total transcripts is protein-coding [17]; the rest of the transcripts are non-coding RNAs (ncRNAs). There are two major classes of regulatory ncRNAs based on their size: small ncRNAs (< 200 nt in length) represented by micro- RNAs, and long ncRNAs (lncRNAs) (> 200 nt in length) [18,19]. LncRNAs are classified into six groups, based on genomic organization relative to protein-coding genes (PCGs), which is summarized in Fig. 2. There are a large number of lncRNAs reported in different databases, such as the encode database (http://www.encode.com), noncode database (http://www.noncode.org), and LNCipedia (http://www.lncipedia.org). The number of reported lncRNA ranges from < 20,000 to over 100,000 in humans [20]. The large variation in the number of lncRNAs among databases results from the type of annotation method, whether automated and/or manual. Both types of annotations suffer from tradeoffs between quality and size, and are comprehensively reviewed by Uszczynska-Ratajczak et al. [21]. LncRNAs display several similarities to PCGs; the majority of lncRNAs are transcribed by Pol II, and harbor histone modifications associated with Pol II transcriptional elongation [22]. Furthermore, lncRNAs are 5′-capped [23] and many are polyadenylated [24]. Unlike PCGs, lncRNA do not always display evolutionary sequence conservation [25] and most have limited protein-coding potential [26]. Another distinctive feature of lncRNA is tissue- and cell type-specific expression [27]. This feature is clearly evident from studies in our laboratory focusing on lncRNAs in the mouse kidney, in which a small fraction of the annotated murine lncRNA (< 10%) were expressed in the adult mouse kidney [28]. Until recently, lncRNAs were considered to be transcriptional noise; however, it has become increasingly evident that many lncRNAs are involved in numerous biological functions and pathological processes, including development, proliferation, apoptosis, survival, differentiation, and many malignancies [29–32]. LncRNAs were found to influence cellular activities and biological pathways in different mechanisms. For example, some lncRNAs regulate gene transcription by interacting with transcriptional complexes, DNA elements, and chromatin structures [33], while others function post-transcriptionally by regulating mRNA stability, splicing and modifications, as well as protein translation [34]. lncRNA function, in part, can be deduced from the cellular localization of the transcript. For example, MALAT1, a well-known nuclear lncRNA, is involved in the assembly of nuclear speckles and transcriptional regulation of metastatic genes [35–37]. NORAD, on the other hand, is abundant in cytoplasm, and regulate genomic stability by sequestering PUMILIO proteins that repress the stability and translation of mRNAs to which they bind [38,39].
Fig. 2.
Positional classification of lncRNAs relative to nearest protein-coding-gene (PCG). Intronic lncRNAs are found within an intron of a PCG, whereas exonic lncRNAs overlap with an exon of a PCG. Both can be transcribed in a sense or anti-sense direction. Bidirectional lncRNAs share the same promoter of nearby PCG. Intergenic lncRNAs donot overlap with a PCG.
This review will summarize the latest findings that implicate lncRNAs in the regulation of mTOR signaling. We will provide a detailed description of lncRNAs that regulate mTOR signaling in an identified mechanism of action, and briefly discuss other lncRNAs that influence mTOR signaling in various conditions and diseases. We will then discuss the significance of these findings in the context of lncRNAs as potential therapeutic targets.
2. lncRNAs and mTOR signaling
In the past decade, many studies have reported dysregulation of mTOR signaling in response to altered expression of lncRNA and vice versa [40]; however, only a small number of these studies describe the mechanism of regulation. For example, some lncRNAs influence mTOR activity by directly binding to components of the mTOR complex, while others regulate proteins that are upstream or downstream of mTOR. In addition, some lncRNAs function via a lncRNA-miRNA network that affects mTOR signaling. Other studies describe correlative effects between lncRNA expression and mTOR signaling. All the lncRNAs described in this review are listed in Table 1.
Table 1.
mTOR-Associated lncRNAs.
lncRNA | Expression | Disease | Effect on mTOR | Mechanism | Reference |
DLEU1 | Upregulated | Endometrial cancer | Activation | Binds mTOR complex | Du et al., 2018 [46] |
HAGLROS | Upregulated | Gastric cancer | Activation | 1- Interacts with mTOR, Raptor, and PRAS40 2- Antagonize miR-100 |
Chen et al., 2018 [47] |
NBR2 | Downregulated | Cancer cells | Suppression | Binds to AMPK | Liu et al., 2016 [49] |
H19 | Downregulated | Pituitary cancer | Suppression | Binds to and prevents 4E-BP1 phosphorylation | Wu et al., 2018 [56] |
FA2H-2 | Downregulated | Atherosclerosis | Suppression | Represses MLKL transcription | Guo et al., 2019 [57] |
LINC00152 | Upregulated | Hepatic cancer | Activation | Increases EpCAM transcription | Ji et al., 2015 [61] |
MALAT1 | Upregulated | Hepatic cancer | Activation | Increases expression of S6K1-isoform 2 | Malakar et al., 2017[68] |
MetaLnc9 | Upregulated | Non-small cell lung cancer | Activation | Binds to and stabilizes PGK1 expression | Yu et al., 2017 [69] |
Dlk1-Gtl2 locus | Downregulated | Normal liver development | Suppression | Give rise to miRNA cluster that targets mTOR pathway | Qian et al., 2016[72] |
BFAL | Upregulated | Gut microbiota-induced colorectal cancer | Activation | Regulates RHEB expression by sponging miR-155–5p and miR-200a-3p | Bao et al., 2019 [73] |
DANCR | Upregulated | Lung cancer | Activation | Antagonize miR-496 | Lu et al., 2018 [80] |
ANRIL | Upregulated | Gastric cancer | Activation | Repression of miR-99a and miR-449a | Zhang et al., 2014 [83] |
GAS5 | Downregulated | Multiple cancers | Repression | Mourtada-Maarabouni et al., 2009 [87] Pickard et al., 2013 [88] Cao et al., 2014 [89] Sun et al., 2014 [90] |
|
Hoxb3os | Downregulated | Polycystic kidney disease | Suppression | Aboudehen et al., 2018 [28] | |
CRNDE | Upregulated | Glioma | Activation | Wang et al., 2015 [93] | |
UCA1 | Upregulated | Bladder cancer | Activation | Li et al., 2014[95] | |
NEAT1 | Upregulated | 1) Diabetic Nephropathy 2) Nonalcoholic fatty liver disease |
Activation | Huang et al., 2019 [98] Wang et al., 2018 [77] | |
HOTAIR | Upregulated | 1) Hepatic cancer 2) Intervertebral degeneration |
Activation Suppression |
Wei et al., 2017 [102] Yang et al., 2019 [103] Zhan et al., 2019 [104] | |
ZNNT1 | Downregulated | Uveal melanoma | Suppression | Li et al., 2019 [105] | |
HULC | Upregulated | Human Gliomas | Activation | Zhu et al., 2016 [110] |
2.1. lncRNAs that directly interacts with mTOR complex
Two lncRNAs physically interact with the mTOR complex: DLEU1 and HAGLROS. In both studies, lncRNA binding to the mTOR complex was determined by RNA immunoprecipitation. Thus, additional experiments, such as RNA/protein crosslinking-based methods, are needed to validate and determine the level of interaction.
-DLEU1 (Deleted in lymphocytic leukemia 1)
This lncRNA is located at the 13q14.3 in humans, a chromosomal region that is recurrently deleted in B-cell chronic lymphocytic leukemia (CLL) [41]. Because 13q14 is a known tumor suppressor locus that contains the Retinoblastoma (RB1) gene, DLEU1 was thought to be a tumor suppressor gene. However, the DLEU1 prompter is hyperactive in leukemia cells and causes silencing of nearby tumor suppressor genes [42]. In addition, DLEU1 functions as an oncogene in various carcinomas including colorectal, gastric, ovarian, and oral [43–45]. Evidence implicating DLEU1 in mTOR signaling is described in endometrial cancer, in which DLEU1 levels are elevated in patients with the disease compared to healthy subjects [46]. In vitro overexpression of DLEU1 results in increased cell viability, migration, invasion, and decreased apoptosis, whereas DLEU1 knockdown produces the opposite effects [46]. At the molecular level, DLEU1 overexpression results in increased mTOR phosphorylation and activation of downstream targets, including pS70K. RNA immunoprecipitation (RIP) assays reveal that DLEU1 is immunoprecipitated with an antibody against mTOR protein. The results of this study indicate that DLEU1 enhances tumorigenesis by binding mTOR protein and activating mTORC1.
-HAGLROS (HAGLR opposite strand)
Is a lncRNA that localizes to the HoxD cluster of homeotic genes and overlaps with another lncRNA called HAGLR (HoxD antisense growth-associated long-oncoding RNA) [47]. Unlike HAGLR, which is involved in the postnatal growth and development of the mouse, little is known about the role and function of HAGLROS. Recent studies reveal that overexpression of HAGLROS contributes to the malignancy of colorectal and gastric cancer cells by decreasing cell apoptosis and autophagy [48]. In gastric cancer, HAGLROS is activated as a result of increased binding of STAT3 transcription factor to the promoter region [47]. HAGLROS activation increases total and phospho-mTOR and decreases autophagy. Mechanistically, HAGLROS activates mTOR signaling and inhibits autophagy in dual manner. On one hand, HAGLROS functions as a competing endogenous RNA (CeRNA) that antagonizes miR100–5p-mediated degradation of mTOR. On the other hand, HAGLROS interacts directly with mTOR, Raptor, and PRAS40, which stabilizes mTORC1 activity [47].
2.2. lncRNAs that function upstream or downstream of mTOR signaling
A number of lncRNAs modulates mTOR pathway by regulating molecules that are upstream and downstream of mTOR complex (Fig. 3). These lncRNAs include:
Fig. 3.
lncRNAs that function upstream and downstream of mTOR signaling.
-NBR2 (Neighbor of BRCA1 gene 2)
Liu et al. identified NBR2 as a glucose starvation-induced lncRNA in various cancer cell lines, excluding Hela or A549 cells, which lack the tumor suppressor liver kinase B1 (LKB1) gene [49]. Re-expression of Lkb1 restores energy stress-induced NBR2, while AMPK inactivation lowers NBR2 induction. The data indicate that NBR2 function through the LKB1-AMPK pathway. Further analysis reveals a NBR2-AMPK feedback-forward loop, in which AMPK induces NBR2 expression and NBR2, in turn, potentiates AMPK activity. RNA-pulldown assays reveal that NBR2 interacts with AMPKα subunit and the interaction significantly increases in response to glucose starvation. AMPK is an upstream negative regulator of mTORC1 signaling that acts by phosphorylating Raptor and the TSC1-TSC2 complex. AMPK also promotes autophagy through direct phosphorylation of the autophagy regulator ULK1 [50,51]. As a result, the investigators examined whether mTOR signaling and cell autophagy are influenced by NBR2 activity. NBR2 knockdown rendered cancer cells partially resistant to energy stress-induced mTORC1 inactivation, while NBR2 overexpression repressed mTORC1 activity. In addition, energy stress-induced autophagy was defective in NBR2 deficient cells. Results of the study indicate that NBR2 represses mTORC1 activity by binding and activating AMPK.
-H19 (Homo sapiens H19 imprinted maternally expressed transcript)
H19 is an imprinted lncRNA that is exclusively expressed from the maternal allele and is in close proximity to the insulin-like growth factor 2 (IGF2), which is produced from the paternal allele [52,53]. The H19 locus also harbors a microRNA, miR-675, which controls placental growth by regulating the expression of the Igf1 gene [54]. Deletion of the H19 locus in the mouse results in an over-growth phenotype that is rescued by overexpression of H19 [55]. Wu et al. found that H19 expression is downregulated in human pituitary adenomas and expression negatively correlates with tumor progression [56]. Restoring H19 expression in tumor cells inhibits cell proliferation and tumor growth. At the molecular level, H19 suppresses 4E-BP1 phosphorylation without affecting S6K1 phosphorylation. To identify the underlying mechanism of H19 specificity towards 4E-BP1 phosphorylation, immunoprecipitation and RNA pulldown assays were performed and reveals that H19 directly binds to 4E-BP1, which prevents Raptor from binding and phosphorylating 4E-BP1.
-FA2H-2:
Guo et al. examined the role of lncRNAs in atherosclerosis by treating cells with oxidized low-density lipoprotein (OX-LDL), a model that induces inflammation and causes atherosclerosis [57]. OX-LDL treatment results in increased expression of the mixed lineage kinase domain-like protein (MLKL), which is known to attenuate autophagy [58], as well as decrased levels of the nearby FA2H-2 lncRNA. In addition, silencing of FA2H-2 or overexpression of MLKL activates inflammation and inhibits autophagy in an mTOR-dependent manner, which suggest that MLKL and FA2H-2 have antagonizing effects on mTOR signaling. Further analysis reveal that FA2H-2 knockdown increases MLKL expression, which indicates that FA2H-2 is an upstream inhibitor of MLKL. This was confirmed by performing Chromatin Isolation by RNA Purification (ChIRP) and luciferase reporter assays, which reveal binding of FA2H-2 to MLKL promoter and repression of the MLKL transcript. The results of this study indicate that FA2H-2 has a therapeutic effect during atherosclerosis by inhibiting MLKL expression and silencing mTOR signaling.
-LINC00152:
This lncRNA was initially identified as a differentially hypomethylated gene in hepatic cancer and is overexpressed in many tumors including lung and gastric cancer [59,60]. Ji et al. found mTOR signaling to be highly active in hepatic cancer cells that overexpress LINC00152 [61]. They also reveal that EpCAM, a gene located near LINC00152, is upregulated in response to LINC00152 overexpression. EpCAM is a type I transmembrane glycoprotein that is overexpressed in a large variety of human epithelial-derived neoplasms, and previous studies describe EpCAM as an upstream inducer of mTOR signaling [62–64]. Luciferase reporter gene assays reveal that knockdown of LINC00152 decreases the promoter activity of EpCAM, and tethering of LINC00152 to the EpCAM reporter stimulates the transcription of the reporter [61]. The results of the study suggest that LINC00152-dependent mTOR hyperactivation results from EpCAM overexpression, whereby LINC00152 binds to and increases EpCAM promoter activity, which causes an increase in mTOR activity.
-MALAT1 (Metastasis-associated lung adenocarcinoma transcript 1):
MALAT1 is one of the most abundantly expressed lncRNA, which was originally identified in metastatic non-small-cell lung cancer cells [65,66]. MALAT1 localizes to nuclear bodies known as nuclear speckles, a region that contains a distinct set of pre-mRNA splicing factor, and specifically interacts with SRSF1, 2, and 3 [35,67]. Malakr et al. reveal that upregulation of MALAT1 in hepatocellular carcinoma causes increased mTOR signaling via effects on the splicing factor SRSF1 [68]. Induction of SRSF1 by MALAT1 modulates SRSF1 splicing targets, one of which is S6K1. In response to activation by MALAT1, SRSF1 favors the formation of S6K1-isoform 2, a shorter spliced variant of S6K1. S6K1-isoform 2 possesses an oncogenic activity that binds and activates mTORC1, which results in increased phosphorylation of 4E-BP1.
-MetaLnc9:
MetaLnc9, also known as LINC00963, is located on chromosome 9 and has one transcript containing five exons [20]. MetaLnc9 is highly abundant in non-small cell lung cancer (NSCLC), and expression correlates with enhanced cell migration and invasion [69]. Yu et al. examined the biological function of MetaLnc9 in NSCLC, both in vitro and in vivo. They found that AKT/mTOR signaling was inhibited by MetaLnc9 knockdown and activated upon MetaLnc9 overexpression [69]. RNA-pulldown assay followed by mass spectrometry revealed that MetaLnc9 interacts with phosphoglycerate kinase 1 (PGK1) protein. This interaction prevents PGK1-mediated ubiquitination, which results in PGK1 accumulation and subsequent mTOR activation. The inhibitory effects of MetaLnc9 knockdown on AKT/mTOR signaling was rescued upon PGK1 overexpression and reversed by PGK1 knockdown. Results of this study demonstrate that MetaLnc9 directly binds to and stabilizes PGK1 protein, which leads to mTOR activation.
2.3. mTOR regulation through lncRNA-miRNA interaction
Studies identify several lncRNAs that influence mTOR signaling through a miRNA/mTOR regulatory network. These lncRNAs function in different mechanisms; for example, the Dlk1-Gtl2 locus harbors many lncRNAs that give rise to multiple miRNAs that target mTOR components. In contrast, BFAL1 and DANCR function as a competitive endogenous RNA (ceRNA) that sequester miRNAs and alleviate the mTOR-mediated repression. Other lncRNAs, such as ANRIL regulate mTOR activity through transcriptional activation or repression of miRNAs known to target mTOR.
-Dlk1-Gtl2 locus:
Is an imprinted locus, which contains three protein-coding genes (Dlk1, Rtl1, and Dio3) on the paternally inherited allele and produces multiple lncRNAs from the maternally inherited allele. These lncRNAs give rise to multiple miRNAs, including the largest miRNA mega-cluster in mammals (anti-Rtl1, which contains the miR-127/miR-136 cluster with 7 miRNAs, and Mirg, which contains the miR-379/miR-410 cluster with 39 miRNAs). miRNAs encoded within the Dlk1-Gtl2 locus have important cellular functions, such as regulation of skeletal muscle differentiation and neurogenesis [70,71]. In hematopoietic stem cells (HSCs), Dlk1-Gtl2 expression is required for the maintenance of HSC population in the mouse fetal liver, and loss of imprinting at the locus results in significant reduction of HSCs due to AKT-mTOR hyperactivation and subsequent metabolic abnormalities [72]. Mechanistically, the miRNA mega-cluster within the Dlk1-Gtl2 locus suppresses the entire PI3K-mTOR pathway. Investigators identified 21 genes that are distributed throughout the PI3K-mTOR pathway and harbors one or multiple miRNA binding sites in their respective 3′ UTRs. Regulation of these targets by the miRNA cluster was confirmed by luciferase reporter and rescue experiments [72].
-BFAL1 (Bacteroides fragilis-associated lncRNA1)
Bao et al. identified BFAL1 as a novel lncRNA in colorectal cancer (CRC) that is caused by the Enterotoxigenic Bacteroides fragilis (ETBF), a known tumor-inducing bacterium in the human gut [73]. Patients with CRC display high levels of BFAL1, which correlates with tumor size and reduce survival. Infection of CRC cells with ETBF upregulates BFAL1, and injection of BFAL1 overexpressing cells into a xenograft mouse model results in increased tumorigenesis due to mTOR hyperactivation. Further analysis reveals that BFAL1 overexpression upregulates RHEB, a Ras homolog that binds and activates the mTOR complex. Mechanistically, BFAL1 upregulates RHEB post transcriptionally by binding and sequestering two miRNAs that normally target RHEB, miR155–5p and miR200a-3p [73].
-DANCR (Anti-differentiation ncRNA)
DANCR was identified as an oncogenic lncRNA in multiple malignant tumors [74,75]. DANCER is a known regulator of multiple miRNAs, [76,77] transcription factors [78], and chromatin modifiers [79]. Lu et al. reveal that DANCR levels are upregulated in lung cancer and inversely correlate with miR-496 expression [80]. Knockdown of DANCR by siRNA increased miR-496 levels and overexpression suppressed miR-496. Investigators utilized two assays to demonstrate mir-496/DANCR interaction. First, miR-496 mimic decreased the luciferase activity of a plasmid that harbors DANCR sequence. Second, a miRNA pulldown assay utilizing biotinylated miR-496 mimic reveals that miR-496 directly interacts with DANCR. Since miR-496 is known to target mTOR [81], investigators tested the hypothesis that DANCR can modulate mTOR signaling via miR-496. In vitro assays reveal that knockdown of DANCR reduced the levels of mTOR mRNA and protein, and the effects are reversed by a miR-496 inhibitor. The results of this study indicate that DANCR modulates mTOR activity by acting as a ceRNA for miR-496.
-ANRIL (Antisense Noncoding RNA in the INK4 locus):
ANRIL is a lncRNA that is transcribed in an antisense direction to the tumor suppressor gene CDKN2B (Cyclin Dependent Kinase Inhibitor 2B). ANRIL regulates CDKN2B expression by interacting with the Polycomb proteins PRC1 and PRC2 [82]. Zhang et al. found that expression of ANRIL is upregulated in patients with gastric cancer, and ANRIL levels negatively correlate with patient survival [83]. In vitro and In vivo assays reveal that silencing of ANRIL results in decreased cell proliferation and viability. At the molecular level, ANRIL represses the expression of miR-99a and miR-449a, two miRNA that are known to target mTOR. This is due to recruitment of the PRC2 complex to the miRNA promoter region, causing a decrease in miR-99a/miR-449a expression and subsequent activation of mTOR.
2.4. lncRNAs dysregulations that correlate with mTOR signaling
Many studies describe dysregulations in lncRNA expression in response to changes in mTOR activity and vice versa; however, the mechanism(s) behind lncRNA/mTOR dysregulations have yet to be determined Some of the lncRNAs are described below.
-GAS5 (Growth arrest-specific transcript 5):
GAS5 was originally identified as a tumor suppressor lncRNA that is induced in response to serum starvation or treatment with rapamycin [84]. Despite having a short open-reading-frame, GAS5 does not possess protein-coding capacity and acts as a host-gene for snoRNAs [85]. The induction of GAS5 transcript is regulated by the interplay between the mTOR and the nonsense-mediated decay (NMD) pathway, whereby GAS5 translation in active mTOR cells is rapidly degraded by the NMD pathway [86]. Thus many cancers, such as breast [87], prostate [88], cervical [89], and gastric [90], have deceased GAS5 expression due to mTOR hyperactivation. GAS5 elicits apoptosis and reduces proliferation by regulating multiple targets, such as c-Myc, Ybx1, cIAP2, and SGK1 [86]. The molecular mechanism by which mTOR inhibition results in GAS5 upregulation is not known.
-Hoxb3os (Homeobox B3 opposite strand):
This lncRNA is located in the HoxB cluster of homeotic genes and is transcribed from the antisense strand. Hoxb3os expression is highly abundant in the kidney and is downregulated in mouse and human kidneys with autosomal dominant polycystic kidney disease (ADPKD) [28]. In situ hybridization assays demonstrate that Hoxb3os transcript is primarily cytoplasmic and localizes to different segments of the nephron. Deletion of Hoxb3os in normal kidney cells results in increased phosphorylation of mTOR protein and activation of downstream targets of mTORC1, including p70S6 kinase, RS6, and 4E-BP1 [28]. Consistent with mTROC1 activation, Hoxb3os-mutant cells display enhanced mitochondrial respiration. The Hoxb3os-mutant phenotype is rescued upon re-expressing Hoxb3os in knockout cells using a lentiviral vector. The results of this study suggest that suppression of Hoxb3os may contribute to activation of mTOR pathway and restoration of Hoxb3os may have therapeutic benefits in ADPKD.
-CRNDE (Colorectal neoplasia differentially expressed):
CRNDE shares a bi-directional promoter with iroquois homeobox 5 (IRX5) gene and gives rise to 10 different transcript variants [91]. CRNDE expression is upregulated in a number of neoplastic diseases, specifically in tissues where the gene is not normally expressed [92]. In glioma cell lines and primary glioma samples from patients, CRNDE transcript variants two and four are upregulated by as much as 50-fold [93]. Overexpression or knockdown of CRNDE affects the growth, migration, and invasion of normal and glioma cells. Interestingly, knockdown of CRNDE does not alter the levels of total or phosphorylated mTOR, but results in increased expression of P70S6K, a downstream mTOR target [93], which suggests that CRNDE can modulate the downstream activity of mTORC1 signaling.
-UCA1 (Urothelial cancer associated 1):
UCA1 was first cloned and identified from a bladder cancer cell line [94]. Studies identified oncogenic functions for UCA1 in various cancers by upregulating mTOR signaling [40]. In bladder cancer cells, UCA1 activates mTOR signaling and results in increased glycolysis, in part due to activation of Hexokinase 2 (HK2), an enzyme that catalyzes the first and irreversible step in the ATP-dependent phosphorylation of glucose to yield glucose-6-phosphate [95]. Two major signals control HK2 expression as a result of mTOR activation: a) STAT3, an mTOR downstream effector, promotes HK2 transcriptional activation and b) mTOR dependent repression of miR-143, which normally targets HK2 mRNA, restores HK2 expression at the post-transcriptional level. Results of this study indicate that UCA1 increases glucose metabolism in cancer cells through the mTOR-STAT3/miR143-HK2 signaling cascade.
- NEAT1 (Nuclear paraspeckle assembly transcript 1):
NEAT1 is adjacent to the MALAT1 locus, and is an essential architectural component of paraspeckle nuclear bodies [96]. NEAT1 is upregulated and plays an oncogenic role in many solid tumor [97]. In diabetic nephropathy (DN), NEAT1 is upregulated and correlates with increased AKT/mTOR signaling, and NEAT1 silencing improves renal function and lowers mTOR activation in vitro [98]. These data imply that NEAT1 contributes to progression of DN by activating Akt/mTOR signaling. Similar findings for NEAT1 were reported in nonalcoholic fatty liver disease (NAFLD), in which NEAT1 expression and mTOR signaling are increased in vivo models of NAFLD [99].
-HOTAIR (HOX transcript antisense RNA):
HOTAIR overlaps with the HoxC genes on chromosome 12 and is transcribed from the negative strand. HOTAIR expression is upregulated in multiple tumors such as liver [100] and breast cancer [101]. Overexpression of HOTAIR in hepatocellular carcinoma cells (HCC) causes increased glucose consumption and lactate production [102]. The HOTAIR-dependent glycolytic flux results from enhanced mTOR signaling, as well as binding to and increasing the expression of glucose transporter isoform 1 (GLUT1). Yang el al. examined the role of HOTAIR in exosomal release during HCC tumorigenesis. Their studies reveal that overexpression of HOTAIR results in increased release of exosomes, which is partly due to mTOR-dependent phosphorylation of synaptosome associated protein 23 (SNAP23) [103]. Other studies reveal that HOTAIR effects on mTOR signaling appears to be cell-type specific. For example, Zhan et al. investigated the effect of HOTAIR in Intervertebral disc degeneration (IDD) utilizing nucleus pulposus cells. Contrary to its role in HCC, HOTAIR overexpression enhances autophagy and promotes apoptosis and senescence of NP cells by increasing p-AMPK and p-ULK1 protein expression, as well as lowering p-mTOR levels [104].
-ZNNT1 (The ZNF706 gene neighboring transcript 1):
ZNNT1 is located on chromosome eight, and contains one exon. Li et al. identified ZNNT1 as a downstream target of mTOR signaling, and is induced in uveal melanoma (UV) cells treated with rapamycin, which suggest that ZNNT1 is a downstream of mTOR signaling [105]. Expression analysis followed by mass spectrometry reveals that ZNNT1 upregulates the expression of ATG12, a ubiquitin-like protein that conjugates to ATG5 to facilitate the lipidation of LC3/Atg8 on autophagosomes [106]. Further experiments demonstrate that ZNNT1-dependent upregulation of ATG12 promotes autophagy by increasing the conversion of LC3-I to LC3-II and SQSTM1 degradation; ZNNT1 inhibition, on the other hand, produced the opposite effects. The mechanism by which mTOR upregulates ZNNT1 expression is not known.
-HULC (Highly up-regulated in liver cancer):
HULC is implicated in various cellular processes and is upregulated in many human malignancies, such as esophageal cancer [107], pancreatic cancer [108] and hepatocellular carcinoma [109]. HULC is overexpressed in patients with gliomas, and silencing of HULC suppresses angiogenesis by inhibiting cells proliferation and invasion [110]. At the molecular level, HULC inhibition decreased the phosphorylation of ERK, AKT, mTOR and the downstream molecule eIF4E, which suggests that HULC is an upstream regulator of mTOR signaling in human gliomas.
3. LncRNAs as therapeutic targets
The development of RNA-targeting therapeutics provides tremendous opportunities to modulate pathogenic molecules and pathways in various diseases. One of the distinctive features of lncRNAs is their highly tissue- and cell type-specific expression which could serve as prognostic markers for diagnosing diseases, or even predicting responses to treatments. In addition, as mentioned in this review, many lncRNAs were found to regulate specific genes and signaling pathways including mTOR. Therefore, drugs that target lncRNAs, such as synthetic antisense oligonucleotides (ASOs), can be less toxic and more refined than conventional protein-targeting drugs [111]. There is an increased focus on developing ASOs for the knockdown of deleterious lncRNAs in wide range of human diseases [112]. One such example is Angelman syndrome, a neurological disorder characterized by severe intellectual and developmental disabilities, has been successfully rescued in mice by inhibiting the anti-sense transcript, Ube3a-ats, which normally represses the imprinted ubiquitin protein ligase E3a gene, Ube3a [113]. Other classes of RNA-based therapeutics are currently being developed including splice-switching oligonucleotides, such as the ASO drug Spinraza™, which is clinically approved for correcting a splice switch in the SMN2 (survival of motor neuron 2) gene in the CNS of patients suffering from spinal muscular atrophy (SMA) [114]. These recent successes with RNA-targeting therapeutics have allowed companies to focus their efforts on developing and improving oligonucleotide chemistry for less toxic, more stable and efficient targeting in vivo. For instance, locked nucleic acids (LNAs) are a new class of ASOs that mediate the degradation, as well as the blockage of lncRNA activity [27]. As of today, there are no clinical trials that target lncRNAs that modulate mTOR signaling; however, clinical trials will become more likely as more lncRNA-mTOR mechanistic studies emerge.
4. Concluding remarks
Over the last decade, there has been an exponential increase in studies that aim to investigate the role of lncRNAs in human diseases. New mechanisms of gene regulations by lncRNAs have been uncovered, and many of the signaling pathways that are dysregulated in human diseases are influenced in a lncRNA-dependent manner. A number of studies have already elucidated the molecular mechanism(s) by which lncRNAs regulate various components of mTOR signaling, and nucleic acid-based therapeutics are emerging as a promising therapeutic approach for modulating lncRNA expression. From a clinical perspective, cell-type and tissue specificity of lncRNAs expression confer significant advantage to selectively modulate mTOR signaling in affected organs without disturbing normal tissues. However, the field is still in its early stages and more research is needed before incorporating lncRNAs into the clinic.
Supplementary Material
Acknowledgements
We thank Kara Eckberg for critically reviewing the manuscript. Research from the author laboratory was supported by the National Institute of Diabetes, Digestive and Kidney Diseases of the National Institutes of Health under award number: KO1-DK116934.
Footnotes
Transparency document
The Transparency document associated with this article can be found, in online version
References
- [1].Saxton RA, Sabatini DM, mTOR signaling in growth, metabolism, and disease, Cell 168 (2017) 960–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Inoki K, Corradetti MN, Guan KL, Dysregulation of the TSC-mTOR pathway in human disease, Nat. Genet. 37 (2005) 19–24. [DOI] [PubMed] [Google Scholar]
- [3].Tee AR, Blenis J, mTOR, translational control and human disease, Semin. Cell Dev. Biol 16 (2005) 29–37. [DOI] [PubMed] [Google Scholar]
- [4].Zarogoulidis P, Lampaki S, Turner JF, Huang H, Kakolyris S, Syrigos K, Zarogoulidis K, mTOR pathway: a current, up-to-date mini-review (review), Oncol. Lett. 8 (2014) 2367–2370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Loewith R, Jacinto E, Wullschleger S, Lorberg A, Crespo JL, Bonenfant D, Oppliger W, Jenoe P, Hall MN, Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control, Mol. Cell 10 (2002) 457–468. [DOI] [PubMed] [Google Scholar]
- [6].Saucedo LJ, Gao X, Chiarelli DA, Li L, Pan D, Edgar BA, Rheb promotes cell growth as a component of the insulin/TOR signalling network, Nat. Cell Biol 5 (2003) 566–571. [DOI] [PubMed] [Google Scholar]
- [7].Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM, The rag GTPases bind raptor and mediate amino acid signaling to mTORC1, Science 320 (2008) 1496–1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Dennis PB, Jaeschke A, Saitoh M, Fowler B, Kozma SC, Thomas G, Mammalian TOR: a homeostatic ATP sensor, Science 294 (2001) 1102–1105. [DOI] [PubMed] [Google Scholar]
- [9].Laplante M, Sabatini DM, mTOR signaling in growth control and disease, Cell 149 (2012) 274–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Ma XM, Blenis J, Molecular mechanisms of mTOR-mediated translational control, Nat. Rev. Mol. Cell Biol 10 (2009) 307–318. [DOI] [PubMed] [Google Scholar]
- [11].Zinzalla V, Stracka D, Oppliger W, Hall MN, Activation of mTORC2 by association with the ribosome, Cell 144 (2011) 757–768. [DOI] [PubMed] [Google Scholar]
- [12].Kazyken D, Magnuson B, Bodur C, Acosta-Jaquez HA, Zhang D, Tong X, Barnes TM, Steinl GK, Patterson NE, Altheim CH, Sharma N, Inoki K, Cartee GD, Bridges D, Yin L, Riddle SM, Fingar DC, AMPK directly activates mTORC2 to promote cell survival during acute energetic stress, Sci. Signal 12 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY, Moffat J, Brown M, Fitzgerald KJ, Sabatini DM, Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1, Dev. Cell 11 (2006) 859–871. [DOI] [PubMed] [Google Scholar]
- [14].Corradetti MN, Guan KL, Upstream of the mammalian target of rapamycin: do all roads pass through mTOR? Oncogene 25 (2006) 6347–6360. [DOI] [PubMed] [Google Scholar]
- [15].Tsang CK, Qi H, Liu LF, Zheng XF, Targeting mammalian target of rapamycin (mTOR) for health and diseases, Drug Discov. Today 12 (2007) 112–124. [DOI] [PubMed] [Google Scholar]
- [16].Thoreen CC, Sabatini DM, Rapamycin inhibits mTORC1, but not completely, Autophagy 5 (2009) 725–726. [DOI] [PubMed] [Google Scholar]
- [17].Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Roder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigo R, Gingeras TR, Landscape of transcription in human cells, Nature 489 (2012) 101–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].St Laurent G, Wahlestedt C, Kapranov P, The landscape of long noncoding RNA classification, Trends Genet. 31 (2015) 239–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, Hertel J, Hackermuller J, Hofacker IL, Bell I, Cheung E, Drenkow J, Dumais E, Patel S, Helt G, Ganesh M, Ghosh S, Piccolboni A, Sementchenko V, Tammana H, Gingeras TR, RNA maps reveal new RNA classes and a possible function for pervasive transcription, Science 316 (2007) 1484–1488. [DOI] [PubMed] [Google Scholar]
- [20].Zhao Y, Li H, Fang S, Kang Y, Wu W, Hao Y, Li Z, Bu D, Sun N, Zhang MQ, Chen R, NONCODE 2016: an informative and valuable data source of long noncoding RNAs, Nucleic Acids Res. 44 (2016) D203–D208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Uszczynska-Ratajczak B, Lagarde J, Frankish A, Guigo R, Johnson R, Towards a complete map of the human long non-coding RNA transcriptome, Nat. Rev. Genet 19 (2018) 535–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Denoeud F, Kapranov P, Ucla C, Frankish A, Castelo R, Drenkow J, Lagarde J, Alioto T, Manzano C, Chrast J, Dike S, Wyss C, Henrichsen CN, Holroyd N, Dickson MC, Taylor R, Hance Z, Foissac S, Myers RM, Rogers J, Hubbard T, Harrow J, Guigo R, Gingeras TR, Antonarakis SE, Reymond A, Prominent use of distal 5′ transcription start sites and discovery of a large number of additional exons in ENCODE regions, Genome Res. 17 (2007) 746–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, Cabili MN, Jaenisch R, Mikkelsen TS, Jacks T, Hacohen N, Bernstein BE, Kellis M, Regev A, Rinn JL, Lander ES, Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals, Nature 458 (2009) 223–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Ozsolak F, Kapranov P, Foissac S, Kim SW, Fishilevich E, Monaghan AP, John B, Milos PM, Comprehensive polyadenylation site maps in yeast and human reveal pervasive alternative polyadenylation, Cell 143 (2010) 1018–1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, Lagarde J, Veeravalli L, Ruan X, Ruan Y, Lassmann T, Carninci P, Brown JB, Lipovich L, Gonzalez JM, Thomas M, Davis CA, Shiekhattar R, Gingeras TR, Hubbard TJ, Notredame C, Harrow J, Guigo R, The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression, Genome Res. 22 (2012) 1775–1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Kopp F, Mendell JT, Functional classification and experimental dissection of long noncoding RNAs, Cell 172 (2018) 393–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Huarte M, The emerging role of lncRNAs in cancer, Nat. Med. 21 (2015) 1253–1261. [DOI] [PubMed] [Google Scholar]
- [28].Aboudehen K, Farahani S, Kanchwala M, Chan SC, Avdulov S, Mickelson A, Lee D, Gearhart MD, Patel V, Xing C, Igarashi P, Long noncoding RNA Hoxb3os is dysregulated in autosomal dominant polycystic kidney disease and regulates mTOR signaling, J. Biol. Chem. 293 (2018) 9388–9398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Lalevee S, Feil R, Long noncoding RNAs in human disease: emerging mechanisms and therapeutic strategies, Epigenomics 7 (2015) 877–879. [DOI] [PubMed] [Google Scholar]
- [30].Wapinski O, Chang HY, Long noncoding RNAs and human disease, Trends Cell Biol. 21 (2011) 354–361. [DOI] [PubMed] [Google Scholar]
- [31].Long J, Badal SS, Ye Z, Wang Y, Ayanga BA, Galvan DL, Green NH, Chang BH, Overbeek PA, Danesh FR, Long noncoding RNA Tug1 regulates mitochondrial bioenergetics in diabetic nephropathy, J. Clin. Invest. 126 (2016) 4205–4218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Ignarski M, Islam R, Muller RU, Long non-coding RNAs in kidney disease, Int. J. Mol. Sci 20 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Ulitsky I, Bartel DP, lincRNAs: genomics, evolution, and mechanisms, Cell 154 (2013) 26–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Peng WX, Koirala P, Mo YY, LncRNA-mediated regulation of cell signaling in cancer, Oncogene 36 (2017) 5661–5667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, Blencowe BJ, Prasanth SG, Prasanth KV, The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation, Mol. Cell 39 (2010) 925–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].West JA, Davis CP, Sunwoo H, Simon MD, Sadreyev RI, Wang PI, Tolstorukov MY, Kingston RE, The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites, Mol. Cell 55 (2014) 791–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, Revenko A, Arun G, Stentrup M, Gross M, Zornig M, MacLeod AR, Spector DL, Diederichs S, The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells, Cancer Res. 73 (2013) 1180–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Lee S, Kopp F, Chang TC, Sataluri A, Chen B, Sivakumar S, Yu H, Xie Y, Mendell JT, Noncoding RNA NORAD regulates genomic stability by sequestering PUMILIO proteins, Cell 164 (2016) 69–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Elguindy MM, Kopp F, Goodarzi M, Rehfeld F, Thomas A, Chang TC, Mendell JT, PUMILIO, but not RBMX, binding is required for regulation of genomic stability by noncoding RNA NORAD, Elife 8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Huang T, Wang M, Huang B, Chang A, Liu F, Zhang Y, Jiang B, Long noncoding RNAs in the mTOR signaling network: biomarkers and therapeutic targets, Apoptosis 23 (2018) 255–264. [DOI] [PubMed] [Google Scholar]
- [41].Garding A, Bhattacharya N, Claus R, Ruppel M, Tschuch C, Filarsky K, Idler I, Zucknick M, Caudron-Herger M, Oakes C, Fleig V, Keklikoglou I, Allegra D, Serra L, Thakurela S, Tiwari V, Weichenhan D, Benner A, Radlwimmer B, Zentgraf H, Wiemann S, Rippe K, Plass C, Dohner H, Lichter P, Stilgenbauer S, Mertens D, Epigenetic upregulation of lncRNAs at 13q14.3 in leukemia is linked to the in Cis downregulation of a gene cluster that targets NF-kB, PLoS Genet. 9 (2013) e1003373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Dal Bo M, Rossi FM, Rossi D, Deambrogi C, Bertoni F, Del Giudice I, Palumbo G, Nanni M, Rinaldi A, Kwee I, Tissino E, Corradini G, Gozzetti A, Cencini E, Ladetto M, Coletta AM, Luciano F, Bulian P, Pozzato G, Laurenti L, Forconi F, Di Raimondo F, Marasca R, Del Poeta G, Gaidano G, Foa R, Guarini A, Gattei V, 13q14 deletion size and number of deleted cells both influence prognosis in chronic lymphocytic leukemia, Genes Chromosomes Cancer 50 (2011) 633–643. [DOI] [PubMed] [Google Scholar]
- [43].Liu T, Han Z, Li H, Zhu Y, Sun Z, Zhu A, LncRNA DLEU1 contributes to colorectal cancer progression via activation of KPNA3, Mol. Cancer 17 (2018) 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Li X, Li Z, Liu Z, Xiao J, Yu S, Song Y, Long non-coding RNA DLEU1 predicts poor prognosis of gastric cancer and contributes to cell proliferation by epigenetically suppressing KLF2, Cancer Gene Ther. 25 (2018) 58–67. [DOI] [PubMed] [Google Scholar]
- [45].Wang LL, Sun KX, Wu DD, Xiu YL, Chen X, Chen S, Zong ZH, Sang XB, Liu Y, Zhao Y, DLEU1 contributes to ovarian carcinoma tumourigenesis and development by interacting with miR-490–3p and altering CDK1 expression, J. Cell. Mol. Med 21 (2017) 3055–3065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Du Y, Wang L, Chen S, Liu Y, Zhao Y, lncRNA DLEU1 contributes to tumorigenesis and development of endometrial carcinoma by targeting mTOR, Mol. Carcinog. 57 (2018) 1191–1200. [DOI] [PubMed] [Google Scholar]
- [47].Chen JF, Wu P, Xia R, Yang J, Huo XY, Gu DY, Tang CJ, De W, Yang F, STAT3-induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal-mediated inhibition of autophagy, Mol. Cancer 17 (2018) 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Zheng Y, Tan K, Huang H, Long noncoding RNA HAGLROS regulates apoptosis and autophagy in colorectal cancer cells via sponging miR-100 to target ATG5 expression, J. Cell. Biochem 120 (2019) 3922–3933. [DOI] [PubMed] [Google Scholar]
- [49].Liu X, Xiao ZD, Han L, Zhang J, Lee SW, Wang W, Lee H, Zhuang L, Chen J, Lin HK, Wang J, Liang H, Gan B, LncRNA NBR2 engages a metabolic checkpoint by regulating AMPK under energy stress, Nat. Cell Biol 18 (2016) 431–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, Shaw RJ, Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy, Science 331 (2011) 456–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Kim J, Kundu M, Viollet B, Guan KL, AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1, Nat. Cell Biol 13 (2011) 132–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Bartolomei MS, Zemel S, Tilghman SM, Parental imprinting of the mouse H19 gene, Nature 351 (1991) 153–155. [DOI] [PubMed] [Google Scholar]
- [53].Tilghman SM, Bartolomei MS, Webber AL, Brunkow ME, Saam J, Leighton PA, Pfeifer K, Zemel S, Parental imprinting of the H19 and Igf2 genes in the mouse, Cold Spring Harb. Symp. Quant. Biol. 58 (1993) 287–295. [DOI] [PubMed] [Google Scholar]
- [54].Bourque DK, Avila L, Penaherrera M, von Dadelszen P, Robinson WP, Decreased placental methylation at the H19/IGF2 imprinting control region is associated with normotensive intrauterine growth restriction but not preeclampsia, Placenta 31 (2010) 197–202. [DOI] [PubMed] [Google Scholar]
- [55].Leighton PA, Ingram RS, Eggenschwiler J, Efstratiadis A, Tilghman SM, Disruption of imprinting caused by deletion of the H19 gene region in mice, Nature 375 (1995) 34–39. [DOI] [PubMed] [Google Scholar]
- [56].Wu ZR, Yan L, Liu YT, Cao L, Guo YH, Zhang Y, Yao H, Cai L, Shang HB, Rui WW, Yang G, Zhang XB, Tang H, Wang Y, Huang JY, Wei YX, Zhao WG, Su B, Wu ZB, Inhibition of mTORC1 by lncRNA H19 via disrupting 4E-BP1/raptor interaction in pituitary tumours, Nat. Commun 9 (2018) 4624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Guo FX, Wu Q, Li P, Zheng L, Ye S, Dai XY, Kang CM, Lu JB, Xu BM, Xu YJ, Xiao L, Lu ZF, Bai HL, Hu YW, Wang Q, The role of the LncRNA-FA2H-2-MLKL pathway in atherosclerosis by regulation of autophagy flux and inflammation through mTOR-dependent signaling, Cell Death Differ. 26 (2019) 1670–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Radonjic-Hoesli S, Wang X, de Graauw E, Stoeckle C, Styp-Rekowska B, Hlushchuk R, Simon D, Spaeth PJ, Yousefi S, Simon HU, Adhesion-induced eosinophil cytolysis requires the receptor-interacting protein kinase 3 (RIPK3)- mixed lineage kinase-like (MLKL) signaling pathway, which is counterregulated by autophagy, J. Allergy Clin. Immunol. 140 (2017) 1632–1642. [DOI] [PubMed] [Google Scholar]
- [59].Zhou J, Zhi X, Wang L, Wang W, Li Z, Tang J, Wang J, Zhang Q, Xu Z, Linc00152 promotes proliferation in gastric cancer through the EGFR-dependent pathway, J. Exp. Clin. Cancer Res. 34 (2015) 135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Chen QN, Chen X, Chen ZY, Nie FQ, Wei CC, Ma HW, Wan L, Yan S, Ren SN, Wang ZX, Long intergenic non-coding RNA 00152 promotes lung adenocarcinoma proliferation via interacting with EZH2 and repressing IL24 expression, Mol. Cancer 16 (2017) 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Ji J, Tang J, Deng L, Xie Y, Jiang R, Li G, Sun B, LINC00152 promotes proliferation in hepatocellular carcinoma by targeting EpCAM via the mTOR signaling pathway, Oncotarget 6 (2015) 42813–42824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Kimura O, Takahashi T, Ishii N, Inoue Y, Ueno Y, Kogure T, Fukushima K, Shiina M, Yamagiwa Y, Kondo Y, Inoue J, Kakazu E, Iwasaki T, Kawagishi N, Shimosegawa T, Sugamura K, Characterization of the epithelial cell adhesion molecule (EpCAM)+ cell population in hepatocellular carcinoma cell lines, Cancer Sci. 101 (2010) 2145–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Yamashita T, Budhu A, Forgues M, Wang XW, Activation of hepatic stem cell marker EpCAM by Wnt-beta-catenin signaling in hepatocellular carcinoma, Cancer Res. 67 (2007) 10831–10839. [DOI] [PubMed] [Google Scholar]
- [64].Ni J, Cozzi P, Hao J, Beretov J, Chang L, Duan W, Shigdar S, Delprado W, Graham P, Bucci J, Kearsley J, Li Y, Epithelial cell adhesion molecule (EpCAM) is associated with prostate cancer metastasis and chemo/radioresistance via the PI3K/Akt/mTOR signaling pathway, Int. J. Biochem. Cell Biol. 45 (2013) 2736–2748. [DOI] [PubMed] [Google Scholar]
- [65].Gutschner T, Hammerle M, Diederichs S, MALAT1 - a paradigm for long noncoding RNA function in cancer, J Mol Med (Berl) 91 (2013) 791–801. [DOI] [PubMed] [Google Scholar]
- [66].Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, Thomas M, Berdel WE, Serve H, Muller-Tidow C, MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer, Oncogene 22 (2003) 8031–8041. [DOI] [PubMed] [Google Scholar]
- [67].Nakagawa S, Ip JY, Shioi G, Tripathi V, Zong X, Hirose T, Prasanth KV, Malat1 is not an essential component of nuclear speckles in mice, RNA 18 (2012) 1487–1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Malakar P, Shilo A, Mogilevsky A, Stein I, Pikarsky E, Nevo Y, Benyamini H, Elgavish S, Zong X, Prasanth KV, Karni R, Long noncoding RNA MALAT1 promotes hepatocellular carcinoma development by SRSF1 Upregulation and mTOR activation, Cancer Res. 77 (2017) 1155–1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Yu T, Zhao Y, Hu Z, Li J, Chu D, Zhang J, Li Z, Chen B, Zhang X, Pan H, Li S, Lin H, Liu L, Yan M, He X, Yao M, MetaLnc9 facilitates lung cancer metastasis via a PGK1-activated AKT/mTOR pathway, Cancer Res. 77 (2017) 5782–5794. [DOI] [PubMed] [Google Scholar]
- [70].Snyder CM, Rice AL, Estrella NL, Held A, Kandarian SC, Naya FJ, MEF2A regulates the Gtl2-Dio3 microRNA mega-cluster to modulate WNT signaling in skeletal muscle regeneration, Development 140 (2013) 31–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Seibt J, Armant O, Le Digarcher A, Castro D, Ramesh V, Journot L, Guillemot F, Vanderhaeghen P, Bouschet T, Expression at the imprinted dlk1- gtl2 locus is regulated by proneural genes in the developing telencephalon, PLoS One 7 (2012) e48675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Qian P, He XC, Paulson A, Li Z, Tao F, Perry JM, Guo F, Zhao M, Zhi L, Venkatraman A, Haug JS, Parmely T, Li H, Dobrowsky RT, Ding WX, Kono T, Ferguson-Smith AC, Li L, The Dlk1-Gtl2 locus preserves LT-HSC function by inhibiting the PI3K-mTOR pathway to restrict mitochondrial metabolism, Cell Stem Cell 18 (2016) 214–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Bao Y, Tang J, Qian Y, Sun T, Chen H, Chen Z, Sun D, Zhong M, Chen H, Hong J, Chen Y, Fang JY, Long noncoding RNA BFAL1 mediates enterotoxigenic Bacteroides fragilis-related carcinogenesis in colorectal cancer via the RHEB/mTOR pathway, Cell Death Dis. 10 (2019) 675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Liu Y, Zhang M, Liang L, Li J, Chen YX, Over-expression of lncRNA DANCR is associated with advanced tumor progression and poor prognosis in patients with colorectal cancer, Int. J. Clin. Exp. Pathol 8 (2015) 11480–11484. [PMC free article] [PubMed] [Google Scholar]
- [75].Jiang N, Wang X, Xie X, Liao Y, Liu N, Liu J, Miao N, Shen J, Peng T, lncRNA DANCR promotes tumor progression and cancer stemness features in osteosarcoma by upregulating AXL via miR-33a-5p inhibition, Cancer Lett. 405 (2017) 46–55. [DOI] [PubMed] [Google Scholar]
- [76].Wang S, Jiang M, The long non-coding RNA-DANCR exerts oncogenic functions in non-small cell lung cancer via miR-758–3p, Biomed. Pharmacother. 103 (2018) 94–100. [DOI] [PubMed] [Google Scholar]
- [77].Wang Y, Lu Z, Wang N, Feng J, Zhang J, Luan L, Zhao W, Zeng X, Long noncoding RNA DANCR promotes colorectal cancer proliferation and metastasis via miR-577 sponging, Exp. Mol. Med. 50 (2018) 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Lu Y, Hu Z, Mangala LS, Stine ZE, Hu X, Jiang D, Xiang Y, Zhang Y, Pradeep S, Rodriguez-Aguayo C, Lopez-Berestein G, DeMarzo AM, Sood AK, Zhang L, Dang CV, MYC targeted long noncoding RNA DANCR promotes cancer in part by reducing p21 levels, Cancer Res. 78 (2018) 64–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Li Z, Hou P, Fan D, Dong M, Ma M, Li H, Yao R, Li Y, Wang G, Geng P, Mihretab A, Liu D, Zhang Y, Huang B, Lu J, The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer, Cell Death Differ. 24 (2017) 59–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Lu QC, Rui ZH, Guo ZL, Xie W, Shan S, Ren T, LncRNA-DANCR contributes to lung adenocarcinoma progression by sponging miR-496 to modulate mTOR expression, J. Cell. Mol. Med. 22 (2018) 1527–1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Rubie C, Kolsch K, Halajda B, Eichler H, Wagenpfeil S, Roemer K, Glanemann M, microRNA-496 - a new, potentially aging-relevant regulator of mTOR, Cell Cycle 15 (2016) 1108–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Yap KL, Li S, Munoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, Gil J, Walsh MJ, Zhou MM, Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a, Mol. Cell 38 (2010) 662–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Zhang EB, Kong R, Yin DD, You LH, Sun M, Han L, Xu TP, Xia R, Yang JS, De W, Chen J , Long noncoding RNA ANRIL indicates a poor prognosis of gastric cancer and promotes tumor growth by epigenetically silencing of miR-99a/miR- 449a, Oncotarget 5 (2014) 2276–2292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Schneider C, King RM, Philipson L, Genes specifically expressed at growth arrest of mammalian-cells, Cell 54 (1988) 787–793. [DOI] [PubMed] [Google Scholar]
- [85].Bassett AR, Akhtar A, Barlow DP, Bird AP, Brockdorff N, Duboule D, Ephrussi A, Ferguson-Smith AC, Gingeras TR, Haerty W, Higgs DR, Miska EA, Ponting CP, Considerations when investigating lncRNA function in vivo, Elife 3 (2014) e03058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Ma C, Shi X, Zhu Q, Li Q, Liu Y, Yao Y, Song Y, The growth arrest-specific transcript 5 (GAS5): a pivotal tumor suppressor long noncoding RNA in human cancers, Tumour Biol. 37 (2016) 1437–1444. [DOI] [PubMed] [Google Scholar]
- [87].Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT, GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer, Oncogene 28 (2009) 195–208. [DOI] [PubMed] [Google Scholar]
- [88].Pickard MR, Mourtada-Maarabouni M, Williams GT, Long non-coding RNA GAS5 regulates apoptosis in prostate cancer cell lines, Biochimica Et Biophysica Acta-Molecular Basis of Disease 1832 (2013) 1613–1623. [DOI] [PubMed] [Google Scholar]
- [89].Cao SH, Liu WL, Li F, Zhao WP, Qin C, Decreased expression of lncRNA GAS5 predicts a poor prognosis in cervical cancer, Int. J. Clin. Exp. Pathol 7 (2014) 6776–6783. [PMC free article] [PubMed] [Google Scholar]
- [90].Sun M, Jin FY, Xia R, Kong R, Li JH, Xu TP, Liu YW, Zhang EB, Liu XH, De W, Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer, BMC Cancer 14 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Graham LD, Pedersen SK, Brown GS, Ho T, Kassir Z, Moynihan AT, Vizgoft EK, Dunne R, Pimlott L, Young GP, Lapointe LC, Molloy PL, Colorectal Neoplasia differentially expressed (CRNDE), a novel gene with elevated expression in colorectal adenomas and adenocarcinomas, Genes Cancer 2 (2011) 829–840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Ellis BC, Molloy PL, Graham LD, CRNDE: a Long non-coding RNA involved in cancer, neurobiology, and development, Front. Genet. 3 (2012) 270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Wang Y, Wang Y, Li J, Zhang Y, Yin H, Han B, CRNDE, a long-noncoding RNA, promotes glioma cell growth and invasion through mTOR signaling, Cancer Lett. 367 (2015) 122–128. [DOI] [PubMed] [Google Scholar]
- [94].Wang F, Li X, Xie X, Zhao L, Chen W, UCA1, a non-protein-coding RNA upregulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion, FEBS Lett. 582 (2008) 1919–1927. [DOI] [PubMed] [Google Scholar]
- [95].Li Z, Li X, Wu S, Xue M, Chen W, Long non-coding RNA UCA1 promotes glycolysis by upregulating hexokinase 2 through the mTOR-STAT3/microRNA143 pathway, Cancer Sci. 105 (2014) 951–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, Chess A, Lawrence JB, An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles, Mol. Cell 33 (2009) 717–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Yu X, Li Z, Zheng H, Chan MT, Wu WK, NEAT1: a novel cancer-related long non-coding RNA, Cell Prolif. 50 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Huang S, Xu Y, Ge X, Xu B, Peng W, Jiang X, Shen L, Xia L, Long noncoding RNA NEAT1 accelerates the proliferation and fibrosis in diabetic nephropathy through activating Akt/mTOR signaling pathway, J. Cell. Physiol. 234 (2019) 11200–11207. [DOI] [PubMed] [Google Scholar]
- [99].Wang X, Down-regulation of lncRNA-NEAT1 alleviated the non-alcoholic fatty liver disease via mTOR/S6K1 signaling pathway, J. Cell. Biochem. 119 (2018) 1567–1574. [DOI] [PubMed] [Google Scholar]
- [100].Yang Z, Zhou L, Wu LM, Lai MC, Xie HY, Zhang F, Zheng SS, Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation, Ann. Surg. Oncol. 18 (2011) 1243–1250. [DOI] [PubMed] [Google Scholar]
- [101].Sorensen KP, Thomassen M, Tan QH, Bak M, Cold S, Burton M, Larsen MJ, Kruse TA, Long non-coding RNA HOTAIR is an independent prognostic marker of metastasis in estrogen receptor-positive primary breast cancer, Breast Cancer Res. Treat 142 (2013) 529–536. [DOI] [PubMed] [Google Scholar]
- [102].Wei S, Fan Q, Yang L, Zhang X, Ma Y, Zong Z, Hua X, Su D, Sun H, Li H, Liu Z, Promotion of glycolysis by HOTAIR through GLUT1 upregulation via mTOR signaling, Oncol. Rep. 38 (2017) 1902–1908. [DOI] [PubMed] [Google Scholar]
- [103].Yang L, Peng XQ, Li Y, Zhang XD, Ma YB, Wu CL, Fan Q, Wei SB, Li HY, Liu JG, Long non-coding RNA HOTAIR promotes exosome secretion by regulating RAB35 and SNAP23 in hepatocellular carcinoma, Mol. Cancer 18 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Zhan S, Wang K, Xiang Q, Song Y, Li S, Liang H, Luo R, Wang B, Liao Z, Zhang Y, Yang C, lncRNA HOTAIR upregulates autophagy to promote apoptosis and senescence of nucleus pulposus cells, J. Cell. Physiol. (2019) 1–14, 10.1002/jcp.29129. [DOI] [PubMed] [Google Scholar]
- [105].Li P, He J, Yang Z, Ge S, Zhang H, Zhong Q, Fan X, ZNNT1 long noncoding RNA induces autophagy to inhibit tumorigenesis of uveal melanoma by regulating key autophagy gene expression, Autophagy (2019) 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Otomo C, Metlagel Z, Takaesu G, Otomo T, Structure of the human ATG12~ATG5 conjugate required for LC3 lipidation in autophagy, Nat. Struct. Mol. Biol 20 (2013) 59–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Kang M, Sang Y, Gu H, Zheng L, Wang L, Liu C, Shi Y, Shao A, Ding G, Chen S, Tang W, Yin J, Long noncoding RNAs POLR2E rs3787016 C/T and HULC rs7763881 A/C polymorphisms are associated with decreased risk of esophageal cancer, Tumour Biol. 36 (2015) 6401–6408. [DOI] [PubMed] [Google Scholar]
- [108].Peng W, Gao W, Feng JF, Long noncoding RNA HULC is a novel biomarker of poor prognosis in patients with pancreatic cancer, Med. Oncol 31 (2014). [DOI] [PubMed] [Google Scholar]
- [109].Yang X, Xie X, Xiao YF, Xie R, Hu CJ, Tang B, Li BS, Yang SM, The emergence of long non-coding RNAs in the tumorigenesis of hepatocellular carcinoma, Cancer Lett. 360 (2015) 119–124. [DOI] [PubMed] [Google Scholar]
- [110].Zhu Y, Zhang XB, Qi L, Cai Y, Yang P, Xuan G, Jiang Y, HULC long noncoding RNA silencing suppresses angiogenesis by regulating ESM-1 via the PI3K/Akt/mTOR signaling pathway in human gliomas, Oncotarget 7 (2016) 14429–14440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Bennett CF, Baker BF, Pham N, Swayze E, Geary RS, Pharmacology of antisense drugs, Annu. Rev. Pharmacol. Toxicol 57 (57) (2017) 81–105. [DOI] [PubMed] [Google Scholar]
- [112].Arun G, Diermeier S, Akerman M, Chang KC, Wilkinson JE, Hearn S, Kim Y, MacLeod AR, Krainer AR, Norton L, Brogi E, Egeblad M, Spector DL, Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss, Genes Dev. 30 (2016) 34–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Meng L, Ward AJ, Chun S, Bennett CF, Beaudet AL, Rigo F, Towards a therapy for Angelman syndrome by targeting a long non-coding RNA, Nature 518 (2015) 409–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, Yamashita M, Rigo F, Hung G, Schneider E, Norris DA, Xia ST, Bennett CF, Bishop KM, Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study, Lancet 388 (2016) 3017–3026. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.