Open Access

Research progress on the interaction between long non‑coding RNAs and RNA‑binding proteins to influence the reprogramming of tumor glucose metabolism (Review)

  • Authors:
    • Weizheng Wu
    • Kunming Wen
  • View Affiliations

  • Published online on: July 15, 2022     https://doi.org/10.3892/or.2022.8365
  • Article Number: 153
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As epigenetic regulators, long non‑coding RNAs (lncRNAs) are involved in various important regulatory processes and typically interact with RNA‑binding proteins (RBPs) to exert their core functional effects. An increasing number of studies have demonstrated that lncRNAs can regulate the occurrence and development of cancer through a variety of complex mechanisms and can also participate in tumor glucose metabolism by directly or indirectly regulating the Warburg effect. As one of the metabolic characteristics of tumor cells, the Warburg effect provides a large amount of energy and numerous intermediate products to meet the consumption demands of tumor metabolism, providing advantages for the occurrence and development of tumors. The present review article summarizes the regulatory effects of lncRNAs on the reprogramming of glucose metabolism after interacting with RBPs in tumors. The findings discussed herein may aid in the better understanding of the pathogenesis of malignancies, and may provide novel therapeutic targets, as well as new diagnostic and prognostic markers for human cancers.

Introduction

In recent years, due to advancements being made in research on tumors, the reprogramming of glucose metabolism in tumor cells has gradually become a focus of research. In normal cells, glucose-converted pyruvate enters the tricarboxylic acid cycle (TCA) for oxidative phosphorylation while in an aerobic environment and undergoes glycolysis to produce lactate only during hypoxia. However, tumor cells still prefer the glycolytic pathway for energy even in an aerobic environment (1). This change in the energy pathway provides certain advantages for the occurrence and development of tumors. First, aerobic glycolysis produces more adenosine triphosphate (ATP) than oxidative phosphorylation for consumption by tumor cells when the substrate is sufficient and the recycling efficiency is sufficiently high (1,2). Second, aerobic glycolysis can improve cellular tolerance to hypoxia and avoid oxidative phosphorylation-induced apoptosis (3). In addition, glycolytic intermediates can also provide the raw materials for anabolism, and the increased production of lactic acid can also decompose and destroy the cellular matrix around tumor cells, which promotes tumor cell migration (4). The scientist, Otto Warburg, discovered the phenomenon of aerobic glycolysis in tumor cells as early as the 1920s and defined it as the Warburg effect (5).

For tumor cells to carry out the Warburg effect, two necessary conditions must be met. On the one hand, due to the increased consumption of glucose by tumor cells, greater amounts of glucose need to be taken up from the extracellular environment. On the other hand, there are numerous enzymatic reactions in the glycolytic pathway, and these glycolytic enzymes play a crucial regulatory role. Therefore, the regulation of these two aspects has become critical for the regulation of the Warburg effect.

Glucose transporters (GLUTs) have been reported to enable glucose to cross hydrophobic cell membranes into cells, thereby mediating the use of glucose (6). Previous research has found that the upregulation of GLUTs promotes the Warburg effect in multiple cancer types (7). Other studies have indicated that the transcriptional upregulation of glycolytic enzymes is closely related to the occurrence of tumors, and that changes in their activity and stability have a direct impact on the Warburg effect (810). In addition, the abnormal expression of certain upstream molecules of GLUTs and glycolytic enzymes will affect them to a certain extent, thereby indirectly regulating the Warburg effect (11).

Long non-coding RNAs (lncRNAs) can participate in tumor metabolism by regulating the metabolic remodeling of various substances, including the metabolic reprogramming of glucose (12,13). lncRNAs refer to a class of RNAs whose transcripts are >200 nucleotides in length and have limited protein-coding capacity; they are one of the most widely studied classes of non-coding RNAs (14). It is necessary for lncRNAs to interact with other cellular macromolecules, such as DNA, proteins and RNA, to drive the cancer phenotype, and their functional effect is to interact with proteins. Through lncRNA-protein interactions, lncRNAs can regulate the functions of proteins directly or regulate protein interactions between other proteins (15). Such RNA-binding and interacting proteins are termed RNA-binding proteins (RBPs), which bind to RNA through one or more globular RNA-binding domains and alter the fate or function of the bound RNA (16). At present, the intricate interaction mechanisms of RNA-RBPs have been reported by a large number of studies (17). The present review article summarizes and discusses the interaction between lncRNAs and RBPs in tumor cells, and their direct or indirect effects on the Warburg effect. Furthermore, the present review article elaborates on the different mechanisms in the cited articles. The topics covered in the present review article are summarized in Figs. 1 and 2, and Table I.

Table I.

lncRNAs interact with RBP to regulate the reprogramming of glucose metabolism in tumor cells.

Table I.

lncRNAs interact with RBP to regulate the reprogramming of glucose metabolism in tumor cells.

A, Regulation by direct action

lncRNARBPTarget of actionResulting effects on cancer(Refs.)
CASC11EIF4A3Enhanced GLUT1 expressionPromotes HCC proliferation, migration and glucose metabolism(19)
Gas6-AS1E2F1Inhibit GLUT1 transcriptionInhibition of LUAD progression and reprogramming of glucose metabolism(20)
HULCLDHA/PKM2Enhanced LDHA/PKM2 phosphorylationPromotes HCC progression and the Warburg effect(44)
CASC8FGFR1Inhibits LDHA phosphorylationInhibits BCa cell growth and glycolysis(45)
CRYBG3LDHAEnhance LDHA activityPromotes LC progression and aerobic glycolysis(46)
CASC9IGF2BP2Improve the stability of HK2mRNAPromote GBM aerobic glycolysis(52)
CDKN2B-AS1IMP3Improve the stability of HK2mRNAPromotes glycolysis and tumor progression in cervical cancer cells(53)
HCG22HURDownregulation of PKM2 mRNA and protein levelsInhibits BCa progression and glycolysis(54)
LINC00470FUSInhibits HK1 degradationPromotes glycolysis in GBM cells and inhibits autophagy(58)
KCNQ1OT1HK2Inhibits HK2 degradationPromote the occurrence and aerobic glycolysis of CRC(62)
AC020978PKM2Inhibits PKM2 degradationPromotes NSCLC cell proliferation and glycolytic pathway(63)
FEZF1AS1PKM2Inhibits PKM2 degradationPromotes CRC proliferation, metastasis and glycolysis(64)
PTCSC3PGK1Promotes PGK1 degradationInhibits PTC glycolysis and cell proliferation(65)
LINC00926STUB1/PGK1Promotes PGK1 degradationInhibition of breast tumor growth, metastasis and Warburg effect(67)
SNHG6hnRNPA1Increase PKM2/PKM1 ratioPromote CRC aerobic glycolysis and tumor development(70)
LNCARODSRSF3Increase PKM2/PKM1 ratioIncreased aerobic glycolysis, malignant transformation and chemoresistance in HCC(72)
SNHG14Lin28APromote PKM2/GLUT1 expressionPromotes aerobic glycolysis and tumorigenesis in glioma(73)
HNF4A-AS1hnRNPUPromote HK2/GLUT1 expressionPromotes NB progression and aerobic glycolysis(77)

B, Regulation by indirect action

lncRNARBPTarget of actionResulting effects on cancer(Refs.)

CASC9HIF-1αIncrease the stability of HIF-1αPromotes glycolysis and tumorigenesis in NPC cells(89)
LINK-ABRK/LRRK2Activates HIF-1α and prevents degradationPromotes glycolytic reprogramming and tumorigenesis in TNBC(90)
lincRNA-p21VHLBlock HIF-1α degradationPromotes tumor growth and glycolysis(91)
GHET1VHLBlock HIF-1α degradationPromotes glycolysis and progression in ovarian cancer cells(92)
EPB41L4A-AS1HDAC2Promote HIF-1α degradationInhibits the Warburg effect of tumor cells(93)
FILNC1AUF1Inhibit c-Myc transcriptionInhibits kidney cancer development and glycolysis(101)
LINC00261IGF2BP1Promotes c-Myc mRNA degradationInhibits glycolysis and proliferation in pancreatic cancer(107)
FGF13-AS1IGF2BP1Promotes c-Myc mRNA degradationInhibits glycolysis and stemness in breast cancer cells(78)
LINRISIGF2BP2Prevent c-Myc mRNA degradationPromotes CRC aerobic glycolysis and proliferation(108)
UCA1UPF1unknownPromotes HCC growth and invasion and glycolysis(109)
ZFAS1IMP2unknownPromotes CRC progression and glycolysis(51)

[i] CASC11, cancer susceptibility candidate 11; GLUT1, glucose transporter 1; HCC, hepatocellular carcinoma; E2F1, E2F transcription factor 1; LUAD, lung adenocarcinoma; HULC, highly upregulated in liver cancer; LDHA, lactate dehydrogenase A; PKM2, pyruvate kinase M2; CASC8, cancer susceptibility candidate 8; FGFR1, fibroblast growth factor receptor 1; BCa, bladder cancer; LC, lung cancer; CASC9, cancer susceptibility candidate 9; IGF2BP2 (IMP2), insulin-like growth factor 2 mRNA binding protein 2; HK2, hexokinase2; GBM, glioblastoma multiforme; IMP3, insulin-like growth factor 2 mRNA binding protein 3; HCG22, HLA complex group 22; HuR, human antigen R; FUS, fused in sarcoma; HK1, hexokinase1; CRC, colorectal cancer; NSCLC, non-small cell lung cancer; FEZF1AS1, FEZF1 antisense RNA 1; PTCSC3, papillary thyroid cancer susceptibility candidate 3; PGK1, phosphoglycerate kinase 1; PTC, papillary thyroid carcinoma; STUB1, STIP1 homology and U-box containing protein 1; SNHG6, small nucleolar RNA host gene 6; hnRNPA1, heterogeneous nuclear ribonucleoprotein A1; PKM1, pyruvate kinase M1; SRSF3, serine-and arginine-rich splicing factor 3; HNF4A-AS1, HNF4A antisense RNA 1; hnRNPU, heterogeneous nuclear ribonucleoprotein U; NB, neuroblastoma; HIF-1α, hypoxia-inducible factor-1α; NPC, nasopharyngeal carcinoma; LINK-A, long-intergenic non-coding RNA for kinase activation; BRK, tyrosine protein kinase 6; LRRK2, leucine-rich repeat kinase 2; TNBC, triple-negative breast cancer; VHL, von Hippel-Lindau; GHET1, gastric carcinoma high expressed transcript 1; HDAC2, histone deacetylase 2; FILNC1, Fox O-induced long non-coding RNA 1; AUF1, adenosine-uridine-rich binding factor 1; LINRIS, long intergenic non-coding RNA for IGF2BP2 stability; UCA1, urothelial cancer associated 1; UPF1, up-frameshift protein 1; ZFAS1, ZNFX1 antisense RNA 1.

lncRNAs interact with RBPs to regulate the reprogramming of glucose metabolism in tumor cells through direct action

GLUTs

Tumor cells have a higher demand for glucose as their metabolic rate is substantially greater than that of normal cells, which require a constant supply of energy and nutrients. It has been demonstrated that the first step in energy metabolism is the uptake of glucose into cells through GLUTs. The gene name of the GLUT family is solute carrier 2 (SLC2A). The family consists of 14 members, of which GLUT1-4 are the four most well-known subtypes, and the expression of GLUT family members is increased in various types of cancer (7). There are nunerous cases of lncRNAs affecting glucose metabolism by regulating GLUT, such as lncRNA HOTAIR promoting glycolysis by upregulating GLUT and activating mammalian target of rapamycin (mTOR) signaling, whereas the knockdown of HOTAIR suppresses this effect (18). However, it was found that when lncRNAs interact with RBPs to modulate the Warburg effect through the modulation of GLUTs, the predominantly regulated isoform is GLUT1 (19,20).

It has been found that lncRNA CASC11 interacts with eukaryotic translation initiation factor 4A3 (EIF4A3) in hepatocellular carcinoma (HCC) and modulates the Warburg effect and tumor progression by affecting the expression of GLUT1 (19). EIF4A3 has been reported to be an important exon junction complex involved in the development of mRNA secondary structure in the 5′untranslated region (UTR) region and promotes the initiation of protein translation (21,22). Mechanistically, the interaction of CASC11 with EIF4A3 leads to an increased expression of the downstream transcription factor E2F1 due to its enhanced mRNA stability, which in turn enhances GLUT1 expression and aerobic glycolysis. Following the silencing of CASC11, GLUT1 expression was found to be downregulated, and tumor progression was also significantly inhibited (19). E2F1 mentioned is an important promoter for cells to enter S phase, can promote the reprogramming of glycolysis and the energy metabolism of tumor cells (23), and plays a critical role in the occurrence and development of various malignant tumors (24,25), as well as being able to function directly as an RBP. In lung adenocarcinoma, lncRNA Gas6-AS1 is downregulated as a tumor suppressor (20). It has been found that Gas6-AS1 directly interacts with E2F1 and downregulates GLUT1 transcription by inhibiting the binding of E2F1 to the GLUT1 promoter region, thereby inhibiting tumor progression and the Warburg effect (20).

Glycolytic enzymes

The activity and stability of glycolytic enzymes directly affect the entire glycolytic metabolic pathway, and they are categorized as rate-limiting enzymes according to whether they can affect the overall speed of the metabolic pathway. Hexokinase (HK) is the first rate-limiting enzyme in aerobic glycolysis, catalyzing the conversion of glucose to glucose-6-phosphate (G-6-P). It has four subtypes, HK1, HK2, HK3 and HK4 (11,26), of which HK2 is more dominant than the other subtypes in promoting aerobic glycolysis (27). The expression level of HK2 has been found to be significantly increased in a variety of cancer tissues, and the silencing of HK2 can effectively reduce the level of aerobic glycolysis and promote the transformation of the metabolic mode of cancer cells to oxidative phosphorylation (2830). Phosphofructokinase-1 (PFK1) is the second rate-limiting enzyme in glycolysis, which catalyzes fructose-6-phosphate (F-6-P) to fructose-1,6-diphosphate (F-1,6-BP) through the use of ATP(31). PFK1 exists as three isoforms (PFK-M, PFK-P and PFK-L) in mammals, and the proportions of these isoforms may vary in tissues with varying metabolic needs (32). Fully activated PFK1 exists as a tetramer, and the formation and stabilization of tetramers largely affects the rate of glycolytic flux (33). The third rate-limiting enzyme in glycolysis is pyruvate kinase (PK), which catalyzes the dephosphorylation of phosphoenolpyruvic acid (PEP) to generate ATP and pyruvate, including the four subtypes of PKM1, PKM2, PKR and PKL (34). Previous research has demonstrated that PKM2 isoforms are highly upregulated in tumor cells and are associated with poor prognosis (35). There are two forms of PKM2: One is a tetramer, which is located in the cytoplasm, has high catalytic activity and can rapidly convert PEP to pyruvate, generating more glycolytic flux and ATP (36), while the other is a monomer or dimer that can translocate into the nucleus, has lower catalytic activity, and acts as a coactivator of several transcription factors (34,37).

In addition, the regulatory effects of enzymes that are not rate-limiting on glycolysis cannot be ignored. Phosphoglycerate kinase (PGK) mainly catalyzes the formation of ATP in the aerobic glycolysis pathway. It has two isoforms: PGK1 and PGK2. PGK1 is ubiquitously expressed in all cells and catalyzes the reversible transfer of phosphate groups from 1,3-bisphosphoglycerate (1,3-BPG) to adenosine diphosphate to generate 3-phosphate glyceric acid (3-PG) and ATP (38). PGK1 is associated with the occurrence and progression of various cancers, and it mediates the ATP produced by glycolysis for use by tumor cells (39). PGK1 and PKM2 are the only two enzymes that control ATP production during aerobic glycolysis in cancer cells (40). The final conversion of pyruvate to lactate depends on lactate dehydrogenase A (LDHA). The abnormal expression and activation of LDHA are closely related to a variety of tumors, and the upregulation of LDHA promotes the malignant progression of tumors by increasing lactate production, accelerating glycolysis, regulating the production of reactive oxygen species, and regulating numerous cancer-related proteins (41).

Glycolytic enzyme activity

It is well known that phosphorylation is a main method of protein post-translational modification. By regulating the degree of protein phosphorylation, the activity of the protein can be significantly affected, thereby participating in the regulation of intracellular biological effects. It has been reported that fibroblast growth factor receptor 1 (FGFR1) can directly phosphorylate LDHA, increasing the binding of LDHA to the substrate nicotinamide adenine dinucleotide dehydrogenase to enhance its enzymatic activity (42), and the intracellular kinase domain of FGFR1 can also phosphorylate PKM2, preventing F-1,6-BP binding to PKM2 and inhibiting active tetramer formation (43). In HCC, lncRNA HULC directly binds and interacts with LDHA and PKM2, and HULC functions as a linker molecule that enhances the binding of LDHA and PKM2 to FGFR1, and increases the phosphorylation levels of LDHA and PKM2 (44). The activity of LDHA is enhanced due to phosphorylation; however, the activity of PKM2 is reduced due to the reduction of the tetrameric form, which ultimately contributes to the Warburg effect and cell proliferation (44). FGFR1 is a membrane receptor of the tyrosine kinase group and can also function as an RBP to interact with lncRNAs to regulate the Warburg effect (45). lncRNA CASC8 exerts a tumor suppressive effect by interacting with FGFR1. Mechanistically, when CASC8 binds to FGFR1, it inhibits the FGFR1-mediated phosphorylation of LDHA and attenuates the conversion of pyruvate to lactate, thereby inhibiting glycolysis and bladder cancer cell growth (45). In addition, it has been demonstrated that lncRNA CRYBG3 interacts with LDHA in lung cancer, and CRYBG3 specifically upregulates the activity of LDHA and promotes aerobic glycolysis and cell proliferation in tumor cells (46).

Glycolytic enzyme stability

In addition to changes in the activity of glycolytic enzymes, certain lncRNAs interact with RBPs to regulate the expression of glycolytic enzymes by affecting the mRNAs of the enzymes. N6-methyladenosine (M6A) is an mRNA modification that can regulate the metabolism of RNAs (mRNAs, miRNAs and lncRNAs) (4749). Insulin-like growth factor 2 mRNA-binding protein (IGF2BP1/2/3, also known as IMP1/2/3) belongs to the K homology domain family and is one of the M6A readers involved in the occurrence and development of cancers by interacting with different RNAs (50,51). lncRNA CASC9 has a M6A site in glioblastoma multiforme (GBM), which is recognized by IGF2BP2 and enhances the stability of CASC9 (52). CASC9 interacts with IGF2BP2 to form an IGF2BP2/CASC9 complex, which can bind to the M6A modification site of HK2, promote the expression of HK2 by improving the stability of HK2 mRNA, and ultimately promote the Warburg effect (52). The circular isoform of lncRNA CDKN2B-AS1, circCDKN2B-AS1, interacts with IMP3 (IGF2BP3) in cervical cancer, and the recruitment of IMP3 (IGF2BP3) to the 3′UTR of HK2 mRNA stabilizes HK2 mRNA and upregulates HK2 expression, promoting a malignant cell phenotype and aerobic glycolysis (53). In addition, lncRNA HCG22 binds and interacts with human antigen R (HUR) in bladder cancer; HUR can positively regulate the expression of polypyrimidine tract-binding protein 1 (PTBP1) (54), and studies have demonstrated that PTBP1 enhances the Warburg effect in certain types of cancer (55,56). When HCG22 is overexpressed, it promotes HUR protein degradation, thereby reducing the expression level of PTBP1 and blocking the Warburg effect mediated by PTBP1, and has been demonstrated that this is achieved by downregulating the mRNA and protein levels of PKM2 (54).

In addition to affecting the state of its mRNA, the stable expression of glycolytic enzymes is also the key to whether or not they will be degraded. It has been reported that >80% of protein degradation is related to the ubiquitin-proteasome pathway (57). For HK, the lncRNA LINC00470 activates protein kinase B (AKT) after interacting with fused in sarcoma protein in GBM (58), and the serine/threonine kinase AKT is a well-established regulator of glucose metabolism that stimulates aerobic glycolysis in tumor cells (5961). These three proteins work together to form a ternary complex. AKT has an enhanced activity due to its increased phosphorylation, and high levels of phosphorylated AKT can inhibit HK1 ubiquitination and attenuate the degradation rate of HK1 protein, thereby affecting the progress of glycolysis (58). In addition, lncRNA KCNQ1OT1 directly binds and interacts with HK2, inhibits the ubiquitination and subsequent degradation of HK2 through the proteasome pathway, and increases the stability of HK2, thereby promoting aerobic glycolysis and cell proliferation in colorectal cancer (CRC) (62). For PKM2, lncRNA AC020978 in non-small cell lung cancer and lncRNA FEZF1-AS1 in CRC both promote tumor progression and aerobic glycolysis, both of which directly bind to PKM2 in mechanism and maintain the stability of PKM2 protein by inhibiting ubiquitin-mediated proteasomal degradation (63,64). The role of PGK1 enzymes has been investigated in papillary thyroid carcinoma (PTC), where the expression of the lncRNA PTCSC3, a tumor suppressor, was found to be significantly downregulated. It was experimentally verified that PTCSC3 directly binds to PGK1, inhibits aerobic glycolysis, and inhibits tumor progression by promoting ubiquitin-mediated degradation of PGK1 (65). Additionally, the STIP1 homology and U-box containing protein 1 (STUB1) has been reported to be an E3 ubiquitin ligase that regulates ubiquitination of multiple substrates (66). In breast cancer cells, STUB1 and PGK1 function together as RBPs to specifically interact with the lncRNA LINC00926, resulting in an enhanced ability of STUB1 to mediate PGK1 ubiquitination, thereby reducing PGK1 expression. LINC00926 inhibits breast tumor growth and metastasis by inhibiting the PGK1-mediated Warburg effect and induces a switch in glucose metabolism from glycolysis to mitochondrial respiration (67).

Glycolytic enzyme isoforms

There are four isoforms of PK, of which PKR and PKL are expressed from the same gene under the control of two different promoters, while PKM1 and PKM2 are generated by the alternative splicing of the PKM gene transcript (68). PKM2 is highly upregulated in cancer cells, and it has been demonstrated that replacing PKM2 with PKM1 can markedly reduce lactate production in tumor cells and the overall tumor size, suggesting that the selection of PKM1 or PKM2 is directly related to the metabolic phenotype of the tumor (69).

Heterogeneous nuclear ribonucleoprotein (hnRNP) is one of the most crucial and classical regulators of alternative splicing. lncRNA SNHG6 is highly expressed in CRC, and it has ben experimentally demonstrated that SNHG6 can interact with hnRNPA1, specifically target the 3′UTR of PKM pre-mRNA, and induce hnRNPA1-specific splicing of PKM pre-mRNA, leading to an increased ratio of PKM2/PKM1, which in turn enhances aerobic glycolysis in CRC cells and promotes tumor development (70). In addition, Ser/Arg-rich splicing factor 3 (SRSF3), known as an alternative splicing regulator, functions as an RBP in various cancer types to play oncogenic roles (71). SRSF3 has been reported to be a PKM splicer that induces the conversion of PKM from PKM1 to PKM2; it induces the conversion of PKM to PKM2 after directly binding to lncRNA LNCAROD in HCC, enhancing aerobic glycolysis, tumorigenesis and chemoresistance in HCC (72).

Regulation of transporters and enzymes simultaneously

In the process of lncRNAs binding to RBPs to regulate the tumor Warburg effect, certain studies have found that GLUT and glycolytic enzymes can be affected simultaneously. lncRNA SNHG14 interacts with RBP Lin28A with an enhanced stability in glioma cells. Stable SNHG14 promotes the degradation of downstream interferon regulatory factor 6 (IRF6) mRNA through the STAU1-mediated degradation pathway and inhibits the expression of IRF6 in cells and tissues, thereby disabling the effect of IRF6, which targets the GLUT1 and PKM2 promoters to repress their expression, and ultimately promoting aerobic glycolysis and cell proliferation in gliomas (73). Among these, IRF6, as a tumor suppressor, has the functions of regulating innate immunity, cell cycle arrest and tumor biological behavior (7476). Additionally, hepatocyte nuclear factor 4 alpha (HNF4A) was identified in neuroblastoma (NB) as a transcription factor that promotes the expression of the glycolytic genes SLC2A1 (GLUT1) and HK2 (77). However, it was found that the expression of HNF4A in NB was regulated by the transcription factor, CCCTC-binding factor (CTCF). Mechanistically, lncRNA HNF4A-AS1 interacts with hnRNPU to promote hnRNPU-mediated CTCF transactivation, and CTCF then increases HNF4A transcription through epigenetic regulation and promotes the Warburg effect and tumor progression through the HNF4A-AS1/hnRNPU/CTCF axis (77).

lncRNAs interact with RBPs to regulate the reprogramming of glucose metabolism in tumor cells through indirect action

Regulation of upstream molecules

In summary, the abnormal expression of GLUT and glycolytic enzymes can directly regulate the Warburg effect in tumor cells. However, there is also the indirect regulation of the Warburg effect by affecting the upstream molecules targeting GLUT and glycolytic enzymes. This chapter focuses on hypoxia inducible factor-1α (HIF-1α) and c-Myc due to their central regulatory roles in cancer cell glycolysis (78).

Due to the rapid proliferation and expansion of cancer cells, there is a phenomenon of hypoxia in the core of tumor tissue, and this abnormal change in the microenvironment will cause continuous metabolic stress on tumor cells. HIF-1α is a transcription factor that is active under specific hypoxic conditions and is a key molecule for cells to adapt to hypoxic conditions (79). It is hydroxylated at proline residues by prolyl hydroxylase under normoxic conditions, and hydroxylated HIF-1α is recognized and bound by von Hippel-Lindau (VHL) proteins that function as ubiquitin E3 ligases, followed by rapid degradation via the ubiquitin-proteasome pathway (8083). However, HIF-1α exhibits an increased stability in hypoxia and binds to the hypoxia response element of target gene promoters, leading to the transcription of related genes involved in overcoming hypoxia effects (32). According to previous research, HIF-1α is mainly involved in the regulation of the Warburg effect in the following aspects. First, HIF-1α can promote the transcription of GLUT1, which promotes glycolysis by enhancing the uptake of glucose by tumor cells (84). Second, HIF-1α can control the transcription of several glycolytic enzymes, such as HK2, PFK1, PKM2, LDHA, etc., by upregulating the expression of these genes, thus further increasing the level of glycolysis (85,86). Finally, HIF-1α can promote the expression of pyruvate dehydrogenase kinase (PDK1) and inhibit pyruvic dehydrogenase (PDH) activity, leading to the conversion of pyruvate to lactate and inhibiting the TCA (87,88). According to previous research, lncRNA CASC9 binds to HIF-1α as an upstream activator of HIF-1α in nasopharyngeal carcinoma, enhancing the stability of HIF-1α and reprogramming cellular glucose metabolism (89). lncRNA LINK-A directly interacts with tyrosine protein kinase 6 (also known as breast tumor kinase) and leucine-rich repeat kinase 2 in triple-negative breast cancer, which activates HIF-1α and prevents its degradation under normoxic conditions, promoting the enhanced Warburg effect (90). In addition, lncRNA lincRNA-p21 and lncRNA GHET1 both block VHL-mediated HIF-1α degradation by interacting with VHL protein in tumors, thereby improving the protein stability and protein level of HIF-1α, and accelerating aerobic glycolysis (91,92). The expression of VHL is also regulated by some indirect mechanisms. It has been reported that lncRNA EPB41L4A-AS1 is a repressor of the Warburg effect and interacts with histone deacetylase 2 (HDAC2). In a previous study, following the knockdown of EPB41L4A-AS1, it was found that the occupancy of HDAC2 on the VHL promoter was increased; histone modification then reduced the expression of VHL and finally activated the HIF-1α pathway to promote glycolysis in tumor cells (93).

c-Myc is the most common oncogene in human carcinogenesis and is involved in the regulation of the cell cycle, cell survival, proliferation and metabolic reprogramming (94). It binds to almost all active promoters and most enhancers, thereby regulating the expression of key genes in the process of cell growth (9597). The regulatory mechanisms of c-Myc in the Warburg effect are similar to those of HIF-1α. Studies have indicated that c-Myc can promote the expression of GLUT1, HK2, PKM2 and LDHA, thereby increasing the glycolytic flux (87,98,99), increasing the cellular uptake of glucose and the production of pyruvate and lactate. c-Myc can also synergistically activate PDK1 with HIF-1α, thereby downregulating PDH and inhibiting the aerobic oxidation of pyruvate, promoting lactate production, and increasing the acidity of the extracellular environment (100). It has been demonstrated that lncRNA FILNC1 is specifically expressed in the kidneys and that FILNC1 interacts with AU-rich elements/poly(U)-binding/degradation factor 1 (AUF1) during energy stress (101). AUF1 has been reported to bind to (A+U)-rich elements within the 3′UTR of c-Myc mRNA and promote its translation without affecting c-Myc mRNA levels (102). FILNC1 reduces c-Myc protein levels by sequestering AUF1 from binding to the c-Myc 3′-UTR; however, this regulatory circuit is dysregulated in renal cancer with a reduced FILNC1 expression (101). Additionally, IGF2BP1/2/3 all physically prevent the degradation of c-Myc mRNA by binding to a consensus sequence containing the ‘GGAC’ M6A core motif (103). lncRNA LINC00261, a widely reported tumor suppressor (104106), interacts with IGF2BP1 in pancreatic cancer and attenuates the c-Myc-mediated Warburg effect by regulating c-Myc mRNA stability (107). Similarly, the interaction of lncRNA FGF13-AS1 with IGF2BP1 in breast cancer shortens the half-life of c-Myc mRNA and ultimately impairs c-Myc-associated glycolysis to inhibit tumor progression (78). lncRNA LINRIS and IGF2BP2 bind and interact in CRC to ensure the stabilization of c-Myc mRNA by preventing the degradation of IGF2BP2, thereby maintaining the c-Myc-mediated Warburg effect and cell proliferation (108).

Unknown target regulatory mechanisms

Among studies on the regulation of glucose metabolism reprogramming following the binding of lncRNAs to RBPs, some did not involve a clear target for regulating the Warburg effect. Rather, changes in glucose consumption, extracellular acidification rates, and lactate production were experimentally demonstrated to explain their effects on the Warburg effect. In HCC tissues, lncRNA UCA1 binds to the tumor suppressor Up-frameshift protein 1 (UPF1), and their expression is inversely correlated. The knockdown of UPF1 has been found to increase the rate of glucose consumption and lactate production (109). In addition, it has been reported that the Obg-like ATPase 1 (OLA1) is an ATP hydrolase that binds to ATP through an amino acid site (110), which plays a crucial role in the production of lactate in tumor cells (111). The stability of lncRNA ZFAS1 is improved after binding to IGF2BP2 in CRC cells, and the binding of stable ZFAS1 to the OBG-type functional domain of OLA1 enhances its ATP hydrolysis ability, thereby promoting the accumulation of lactate and the release of ATP synthesis raw materials, activating the Warburg effect (51).

Conclusions and future perspectives

Glucose metabolic reprogramming is one of the recognized metabolic features of tumor cells, and enhanced glycolysis produces an acidic and hypoxic microenvironment that promotes tumorigenesis, invasion and metastasis. lncRNAs interact with RBPs in a variety of tumor cells, directly or indirectly affecting the Warburg effect by regulating GLUTs, glycolytic enzymes, or their upstream molecules. However, there are still a number of studies that have not yet clearly defined the targets that regulate glycolysis in tumor cells, and further research is still required to elucidate the underlying molecular mechanisms. Interfering with the Warburg effect of tumor cells by affecting the expression of lncRNAs and RBPs or inhibiting GLUTs and glycolytic enzymes can cut off the energy supply, thereby interfering with tumor progression. This provides a potential novel direction and strategy for the targeted therapy of tumors. In the tumor glucose metabolism pathway, there are numerous different molecular targets to choose from to inhibit tumor growth and progression, which may contribute to the advancement of clinical treatment of patients and the improvement of prognosis.

Acknowledgements

Not applicable.

Funding

The present was supported by the National Natural Science Foundation of China (grant no. 82160575) and the Outstanding Young Technological and Innovative Talent Cultivation Project of Zunyi Municipal Science and Technology Bureau, 2021 (no. 10).

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

WW and KW conceived the review. WW was involved in the collection of references. WW wrote the manuscript and constructed the figures. WW and KW checked and revised the manuscript. WW was responsible for the organization, revision and submission of this manuscript. Both authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Vander Heiden MG, Cantley LC and Thompson CB: Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Hsu PP and Sabatini DM: Cancer cell metabolism: Warburg and beyond. Cell. 134:703–707. 2008. View Article : Google Scholar

3 

Kroemer G and Pouyssegur J: Tumor cell metabolism: Cancer's Achilles' heel. Cancer Cell. 13:472–482. 2008. View Article : Google Scholar

4 

Gatenby RA, Gawlinski ET, Gmitro AF, Kaylor B and Gillies RJ: Acid-mediated tumor invasion: A multidisciplinary study. Cancer Res. 66:5216–5223. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Koppenol WH, Bounds PL and Dang CV: Otto Warburg's contributions to current concepts of cancer metabolism. Nat Rev Cancer. 11:325–337. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Mueckler M and Thorens B: The SLC2 (GLUT) family of membrane transporters. Mol Aspects Med. 34:121–138. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Ancey PB, Contat C and Meylan E: Glucose transporters in cancer-from tumor cells to the tumor microenvironment. FEBS J. 285:2926–2943. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Li L, Liang Y, Kang L, Liu Y, Gao S, Chen S, Li Y, You W, Dong Q, Hong T, et al: Transcriptional regulation of the warburg effect in cancer by SIX1. Cancer Cell. 33:368–385.e7. 2018. View Article : Google Scholar

9 

Akins NS, Nielson TC and Le HV: Inhibition of glycolysis and glutaminolysis: An emerging drug discovery approach to combat cancer. Curr Top Med Chem. 18:494–504. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Zheng Y, Liu P, Wang N, Wang S, Yang B, Li M, Chen J, Situ H, Xie M, Lin Y and Wang Z: Betulinic acid suppresses breast cancer metastasis by targeting GRP78-mediated glycolysis and ER stress apoptotic pathway. Oxid Med Cell Longev. 2019:87816902019. View Article : Google Scholar : PubMed/NCBI

11 

Feng J, Li J, Wu L, Yu Q, Ji J, Wu J, Dai W and Guo C: Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res. 39:1262020. View Article : Google Scholar : PubMed/NCBI

12 

Liu C, Li H, Chu F, Zhou X, Xie R, Wei Q, Yang S, Li T, Liang S and Lü M: Long noncoding RNAs: Key regulators involved in metabolic reprogramming in cancer (Review). Oncol Rep. 45:542021. View Article : Google Scholar : PubMed/NCBI

13 

Li Z and Sun X: Non-coding RNAs Operate in the crosstalk between cancer metabolic reprogramming and metastasis. Front Oncol. 10:8102020. View Article : Google Scholar

14 

Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A and Rinn JL: Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25:1915–1927. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar

16 

Hentze MW, Castello A, Schwarzl T and Preiss T: A brave new world of RNA-binding proteins. Nat Rev Mol Cell Biol. 19:327–341. 2018. View Article : Google Scholar

17 

Ferre F, Colantoni A and Helmer-Citterich M: Revealing protein-lncRNA interaction. Brief Bioinform. 17:106–116. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Wei S, Fan Q, Yang L, Zhang X, Ma Y, Zong Z, Hua X, Su D, Sun H, Li H and Liu Z: Promotion of glycolysis by HOTAIR through GLUT1 upregulation via mTOR signaling. Oncol Rep. 38:1902–1908. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Song H, Liu Y, Li X, Chen S, Xie R, Chen D, Gao H, Wang G, Cai B and Yang X: Long noncoding RNA CASC11 promotes hepatocarcinogenesis and HCC progression through EIF4A3-mediated E2F1 activation. Clin Transl Med. 10:e2202020. View Article : Google Scholar : PubMed/NCBI

20 

Luo J, Wang H, Wang L, Wang G, Yao Y, Xie K, Li X, Xu L, Shen Y and Ren B: lncRNA GAS6-AS1 inhibits progression and glucose metabolism reprogramming in LUAD via repressing E2F1-mediated transcription of GLUT1. Mol Ther Nucleic Acids. 25:11–24. 2021. View Article : Google Scholar : PubMed/NCBI

21 

Lu WT, Wilczynska A, Smith E and Bushell M: The diverse roles of the eIF4A family: you are the company you keep. Biochem Soc Trans. 42:166–172. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Chan CC, Dostie J, Diem MD, Feng W, Mann M, Rappsilber J and Dreyfuss G: eIF4A3 is a novel component of the exon junction complex. RNA. 10:200–209. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Wu M, Seto E and Zhang J: E2F1 enhances glycolysis through suppressing Sirt6 transcription in cancer cells. Oncotarget. 6:11252–11263. 2015. View Article : Google Scholar

24 

Chen HZ, Tsai SY and Leone G: Emerging roles of E2Fs in cancer: An exit from cell cycle control. Nat Rev Cancer. 9:785–797. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Farra R, Grassi G, Tonon F, Abrami M, Grassi M, Pozzato G, Fiotti N, Forte G and Dapas B: The role of the transcription factor E2F1 in hepatocellular carcinoma. Curr Drug Deliv. 14:272–281. 2017.PubMed/NCBI

26 

Lis P, Dylag M, Niedzwiecka K, Ko YH, Pedersen PL, Goffeau A and Ułaszewski S: The HK2 dependent ‘Warburg Effect’ and mitochondrial oxidative phosphorylation in cancer: Targets for effective therapy with 3-Bromopyruvate. Molecules. 21:17302016. View Article : Google Scholar

27 

Gong L, Cui Z, Chen P, Han H, Peng J and Leng X: Reduced survival of patients with hepatocellular carcinoma expressing hexokinase II. Med Oncol. 29:909–914. 2012. View Article : Google Scholar

28 

Wolf A, Agnihotri S, Micallef J, Mukherjee J, Sabha N, Cairns R, Hawkins C and Guha A: Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme. J Exp Med. 208:313–326. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Wolf A, Agnihotri S and Guha A: Targeting metabolic remodeling in glioblastoma multiforme. Oncotarget. 1:552–562. 2010. View Article : Google Scholar

30 

Patra KC, Wang Q, Bhaskar PT, Miller L, Wang Z, Wheaton W, Chandel N, Laakso M, Muller WJ, Allen EL, et al: Hexokinase 2 is required for tumor initiation and maintenance and its systemic deletion is therapeutic in mouse models of cancer. Cancer Cell. 24:213–228. 2013. View Article : Google Scholar

31 

Kanai S, Shimada T, Narita T and Okabayashi K: Phosphofructokinase-1 subunit composition and activity in the skeletal muscle, liver, and brain of dogs. J Vet Med Sci. 81:712–716. 2019. View Article : Google Scholar

32 

Al Hasawi N, Alkandari MF and Luqmani YA: Phosphofructokinase: A mediator of glycolytic flux in cancer progression. Crit Rev Oncol Hematol. 92:312–321. 2014. View Article : Google Scholar

33 

Bartrons R, Rodríguez-García A, Simon-Molas H, Castaño E, Manzano A and Navarro-Sabaté À: The potential utility of PFKFB3 as a therapeutic target. Expert Opin Ther Targets. 22:659–674. 2018. View Article : Google Scholar : PubMed/NCBI

34 

van Niekerk G and Engelbrecht AM: Role of PKM2 in directing the metabolic fate of glucose in cancer: A potential therapeutic target. Cell Oncol (Dordr). 41:343–351. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Shang RZ, Qu SB and Wang DS: Reprogramming of glucose metabolism in hepatocellular carcinoma: Progress and prospects. World J Gastroenterol. 22:9933–9943. 2016. View Article : Google Scholar

36 

Anastasiou D, Yu Y, Israelsen WJ, Jiang JK, Boxer MB, Hong BS, Tempel W, Dimov S, Shen M, Jha A, et al: Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat Chem Biol. 8:839–847. 2012. View Article : Google Scholar

37 

Azoitei N, Becher A, Steinestel K, Rouhi A, Diepold K, Genze F, Simmet T and Seufferlein T: PKM2 promotes tumor angiogenesis by regulating HIF-1α through NF-κB activation. Mol Cancer. 15:32016. View Article : Google Scholar : PubMed/NCBI

38 

He Y, Luo Y, Zhang D, Wang X, Zhang P, Li H, Ejaz S and Liang S: PGK1-mediated cancer progression and drug resistance. Am J Cancer Res. 9:2280–2302. 2019.PubMed/NCBI

39 

Daly EB, Wind T, Jiang XM, Sun L and Hogg PJ: Secretion of phosphoglycerate kinase from tumour cells is controlled by oxygen-sensing hydroxylases. Biochim Biophys Acta. 1691:17–22. 2004. View Article : Google Scholar

40 

Hu H, Zhu W, Qin J, Chen M, Gong L, Li L, Liu X, Tao Y, Yin H, Zhou H, et al: Acetylation of PGK1 promotes liver cancer cell proliferation and tumorigenesis. Hepatology. 65:515–528. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Feng Y, Xiong Y, Qiao T, Li X, Jia L and Han Y: Lactate dehydrogenase A: A key player in carcinogenesis and potential target in cancer therapy. Cancer Med. 7:6124–6136. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Fan J, Hitosugi T, Chung TW, Xie J, Ge Q, Gu TL, Polakiewicz RD, Chen GZ, Boggon TJ, Lonial S, et al: Tyrosine phosphorylation of lactate dehydrogenase A is important for NADH/NAD(+) redox homeostasis in cancer cells. Mol Cell Biol. 31:4938–4950. 2011. View Article : Google Scholar

43 

Hitosugi T, Kang S, Vander Heiden MG, Chung TW, Elf S, Lythgoe K, Dong S, Lonial S, Wang X, Chen GZ, et al: Tyrosine phosphorylation inhibits PKM2 to promote the warburg effect and tumor growth. Sci Signal. 2:ra732009. View Article : Google Scholar

44 

Wang C, Li Y, Yan S, Wang H, Shao X, Xiao M, Yang B, Qin G, Kong R, Chen R and Zhang N: Interactome analysis reveals that lncRNA HULC promotes aerobic glycolysis through LDHA and PKM2. Nat Commun. 11:31622020. View Article : Google Scholar : PubMed/NCBI

45 

Hu R, Zhong P, Xiong L and Duan L: Long Noncoding RNA cancer susceptibility candidate 8 suppresses the proliferation of bladder cancer cells via regulating glycolysis. DNA Cell Biol. 36:767–774. 2017. View Article : Google Scholar

46 

Chen H, Pei H, Hu W, Ma J, Zhang J, Mao W, Nie J, Xu C, Li B, Hei TK, et al: Long non-coding RNA CRYBG3 regulates glycolysis of lung cancer cells by interacting with lactate dehydrogenase A. J Cancer. 9:2580–2588. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Dai F, Wu Y, Lu Y, An C, Zheng X, Dai L, Guo Y, Zhang L, Li H, Xu W and Gao W: Crosstalk between RNA m6A Modification and Non-coding RNA Contributes to Cancer Growth and Progression. Mol Ther Nucleic Acids. 22:62–71. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Yang J, Liu J, Zhao S and Tian F: NN 6-Methyladenosine METTL3 modulates the proliferation and apoptosis of lens epithelial cells in diabetic cataract. Mol Ther Nucleic Acids. 20:111–116. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Zhong L, He X, Song H, Sun Y, Chen G, Si X, Sun J, Chen X, Liao W, Liao Y and Bin J: METTL3 Induces AAA development and progression by modulating N6-methyladenosine-dependent primary miR34a processing. Mol Ther Nucleic Acids. 21:394–411. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Wang J, Chen L and Qiang P: The role of IGF2BP2, an m6A reader gene, in human metabolic diseases and cancers. Cancer Cell Int. 21:992021. View Article : Google Scholar

51 

Lu S, Han L, Hu X, Sun T, Xu D, Li Y, Chen Q, Yao W, He M, Wang Z, et al: N6-methyladenosine reader IMP2 stabilizes the ZFAS1/OLA1 axis and activates the Warburg effect: Implication in colorectal cancer. J Hematol Oncol. 14:1882021. View Article : Google Scholar

52 

Liu H, Qin S, Liu C, Jiang L, Li C, Yang J, Zhang S, Yan Z, Liu X, Yang J and Sun X: m 6 A reader IGF2BP2-stabilized CASC9 accelerates glioblastoma aerobic glycolysis by enhancing HK2 mRNA stability. Cell Death Discov. 7:2922021. View Article : Google Scholar : PubMed/NCBI

53 

Zhang Y, Zhao L, Yang S, Cen Y, Zhu T, Wang L, Xia L, Liu Y, Zou J, Xu J, et al: CircCDKN2B-AS1 interacts with IMP3 to stabilize hexokinase 2 mRNA and facilitate cervical squamous cell carcinoma aerobic glycolysis progression. J Exp Clin Cancer Res. 39:2812020. View Article : Google Scholar : PubMed/NCBI

54 

Jiang D, Zhang Y, Yang L, Lu W, Mai L, Guo H and Liu X: Long noncoding RNA HCG22 suppresses proliferation and metastasis of bladder cancer cells by regulation of PTBP1. J Cell Physiol. 235:1711–1722. 2020. View Article : Google Scholar

55 

Minami K, Taniguchi K, Sugito N, Kuranaga Y, Inamoto T, Takahara K, Takai T, Yoshikawa Y, Kiyama S, Akao Y and Azuma H: MiR-145 negatively regulates Warburg effect by silencing KLF4 and PTBP1 in bladder cancer cells. Oncotarget. 8:33064–33077. 2017. View Article : Google Scholar

56 

Taniguchi K, Sakai M, Sugito N, Kumazaki M, Shinohara H, Yamada N, Nakayama T, Ueda H, Nakagawa Y, Ito Y, et al: PTBP1-associated microRNA-1 and −133b suppress the Warburg effect in colorectal tumors. Oncotarget. 7:18940–18952. 2016. View Article : Google Scholar

57 

Wang J and Maldonado MA: The ubiquitin-proteasome system and its role in inflammatory and autoimmune diseases. Cell Mol Immunol. 3:255–261. 2006.

58 

Liu C, Zhang Y, She X, Fan L, Li P, Feng J, Fu H, Liu Q, Liu Q, Zhao C, et al: A cytoplasmic long noncoding RNA LINC00470 as a new AKT activator to mediate glioblastoma cell autophagy. J Hematol Oncol. 11:772018. View Article : Google Scholar

59 

Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, White M, Reichelt J and Levine B: Akt-mediated regulation of autophagy and tumorigenesis through Beclin 1 phosphorylation. Science. 338:956–959. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Le Grand M, Berges R, Pasquier E, Montero MP, Borge L, Carrier A, Vasseur S, Bourgarel V, Buric D, André N, et al: Akt targeting as a strategy to boost chemotherapy efficacy in non-small cell lung cancer through metabolism suppression. Sci Rep. 7:451362017. View Article : Google Scholar : PubMed/NCBI

61 

Polivka J Jr and Janku F: Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol Ther. 142:164–175. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Chen C, Wei M, Wang C, Sun D, Liu P, Zhong X and Yu W: Long noncoding RNA KCNQ1OT1 promotes colorectal carcinogenesis by enhancing aerobic glycolysis via hexokinase-2. Aging (Albany NY). 12:11685–11697. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Hua Q, Mi B, Xu F, Wen J, Zhao L, Liu J and Huang G: Hypoxia-induced lncRNA-AC020978 promotes proliferation and glycolytic metabolism of non-small cell lung cancer by regulating PKM2/HIF-1α axis. Theranostics. 10:4762–4778. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Bian Z, Zhang J, Li M, Feng Y, Wang X, Zhang J, Yao S, Jin G, Du J, Han W, et al: LncRNA-FEZF1-AS1 promotes tumor proliferation and metastasis in colorectal cancer by regulating PKM2 signaling. Clin Cancer Res. 24:4808–4819. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Jiang B, Chen Y, Xia F and Li X: PTCSC3-mediated glycolysis suppresses thyroid cancer progression via interfering with PGK1 degradation. J Cell Mol Med. 25:8454–8463. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Ullah K, Chen S, Lu J, Wang X, Liu Q, Zhang Y, Long Y, Hu Z and Xu G: The E3 ubiquitin ligase STUB1 attenuates cell senescence by promoting the ubiquitination and degradation of the core circadian regulator BMAL1. J Biol Chem. 295:4696–4708. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Chu Z, Huo N, Zhu X, Liu H, Cong R, Ma L, Kang X, Xue C, Li J, Li Q, et al: FOXO3A-induced LINC00926 suppresses breast tumor growth and metastasis through inhibition of PGK1-mediated Warburg effect. Mol Ther. 29:2737–2753. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Chen M, Zhang J and Manley JL: Turning on a fuel switch of cancer: HnRNP proteins regulate alternative splicing of pyruvate kinase mRNA. Cancer Res. 70:8977–8980. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, Fleming MD, Schreiber SL and Cantley LC: The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 452:230–233. 2008. View Article : Google Scholar : PubMed/NCBI

70 

Lan Z, Yao X, Sun K, Li A, Liu S and Wang X: The interaction between lncRNA SNHG6 and hnRNPA1 contributes to the growth of colorectal cancer by enhancing aerobic glycolysis through the regulation of alternative splicing of PKM. Front Oncol. 10:3632020. View Article : Google Scholar

71 

Zhou Z, Gong Q, Lin Z, Wang Y, Li M, Wang L, Ding H and Li P: Emerging roles of SRSF3 as a therapeutic target for cancer. Front Oncol. 10:5776362020. View Article : Google Scholar

72 

Jia G, Wang Y, Lin C, Lai S, Dai H, Wang Z, Dai L, Su H, Song Y, Zhang N, et al: LNCAROD enhances hepatocellular carcinoma malignancy by activating glycolysis through induction of pyruvate kinase isoform PKM2. J Exp Clin Cancer Res. 40:2992021. View Article : Google Scholar : PubMed/NCBI

73 

Lu J, Liu X, Zheng J, Song J, Liu Y, Ruan X, Shen S, Shao L, Yang C, Wang D, et al: Lin28A promotes IRF6-regulated aerobic glycolysis in glioma cells by stabilizing SNHG14. Cell Death Dis. 11:4472020. View Article : Google Scholar : PubMed/NCBI

74 

Ferretti E, Li B, Zewdu R, Wells V, Hebert JM, Karner C, Anderson MJ, Williams T, Dixon J, Dixon MJ, et al: A conserved Pbx-Wnt-p63-Irf6 regulatory module controls face morphogenesis by promoting epithelial apoptosis. Dev Cell. 21:627–641. 2011. View Article : Google Scholar

75 

Rotondo JC, Borghi A, Selvatici R, Magri E, Bianchini E, Montinari E, Corazza M, Virgili A, Tognon M and Martini F: Hypermethylation-Induced inactivation of the IRF6 gene as a possible early event in progression of vulvar squamous cell carcinoma associated with lichen sclerosus. JAMA Dermatol. 152:928–933. 2016. View Article : Google Scholar

76 

Bailey CM, Abbott DE, Margaryan NV, Khalkhali-Ellis Z and Hendrix MJ: Interferon regulatory factor 6 promotes cell cycle arrest and is regulated by the proteasome in a cell cycle-dependent manner. Mol Cell Biol. 28:2235–2243. 2008. View Article : Google Scholar

77 

Song H, Li D, Wang X, Fang E, Yang F, Hu A, Wang J, Guo Y, Liu Y, Li H, et al: HNF4A-AS1/hnRNPU/CTCF axis as a therapeutic target for aerobic glycolysis and neuroblastoma progression. J Hematol Oncol. 13:242020. View Article : Google Scholar

78 

Ma F, Liu X, Zhou S, Li W, Liu C, Chadwick M and Qian C: Long non-coding RNA FGF13-AS1 inhibits glycolysis and stemness properties of breast cancer cells through FGF13-AS1/IGF2BPs/Myc feedback loop. Cancer Lett. 450:63–75. 2019. View Article : Google Scholar

79 

Ruan K, Song G and Ouyang G: Role of hypoxia in the hallmarks of human cancer. J Cell Biochem. 107:1053–1062. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Kaelin WG Jr and Ratcliffe PJ: Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol Cell. 30:393–402. 2008. View Article : Google Scholar

81 

Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A, Asara JM, Lane WS and Kaelin WG Jr: HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing. Science. 292:464–468. 2001. View Article : Google Scholar : PubMed/NCBI

82 

Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, et al: Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 292:468–472. 2001. View Article : Google Scholar : PubMed/NCBI

83 

Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER and Ratcliffe PJ: The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 399:271–275. 1999. View Article : Google Scholar : PubMed/NCBI

84 

Yao J, Man S, Dong H, Yang L, Ma L and Gao W: Combinatorial treatment of Rhizoma Paridis saponins and sorafenib overcomes the intolerance of sorafenib. J Steroid Biochem Mol Biol. 183:159–166. 2018. View Article : Google Scholar

85 

Marín-Hernández A, Gallardo-Pérez JC, Ralph SJ, Rodríguez-Enríquez S and Moreno-Sánchez R: HIF-1alpha modulates energy metabolism in cancer cells by inducing over-expression of specific glycolytic isoforms. Mini Rev Med Chem. 9:1084–1101. 2009. View Article : Google Scholar

86 

Zheng F, Chen J, Zhang X, Wang Z, Chen J, Lin X, Huang H, Fu W, Liang J, Wu W, et al: The HIF-1α antisense long non-coding RNA drives a positive feedback loop of HIF-1α mediated transactivation and glycolysis. Nat Commun. 12:13412021. View Article : Google Scholar : PubMed/NCBI

87 

Kim JW, Tchernyshyov I, Semenza GL and Dang CV: HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 3:177–185. 2006. View Article : Google Scholar : PubMed/NCBI

88 

Yeung SJ, Pan J and Lee MH: Roles of p53, MYC and HIF-1 in regulating glycolysis-the seventh hallmark of cancer. Cell Mol Life Sci. 65:3981–3999. 2008. View Article : Google Scholar

89 

Su X, Li G and Liu W: The long noncoding RNA cancer susceptibility candidate 9 promotes nasopharyngeal carcinogenesis via stabilizing HIF1α. DNA Cell Biol. 36:394–400. 2017. View Article : Google Scholar

90 

Lin A, Li C, Xing Z, Hu Q, Liang K, Han L, Wang C, Hawke DH, Wang S, Zhang Y, et al: The LINK-A lncRNA activates normoxic HIF1α signalling in triple-negative breast cancer. Nat Cell Biol. 18:213–224. 2016. View Article : Google Scholar

91 

Yang F, Zhang H, Mei Y and Wu M: Reciprocal regulation of HIF-1α and lincRNA-p21 modulates the Warburg effect. Mol Cell. 53:88–100. 2014. View Article : Google Scholar

92 

Liu D and Li H: Long non-coding RNA GEHT1 promoted the proliferation of ovarian cancer cells via modulating the protein stability of HIF1α. Biosci Rep. 39:2019.

93 

Liao M, Liao W, Xu N, Li B, Liu F, Zhang S, Wang Y, Wang S, Zhu Y, Chen D, et al: LncRNA EPB41L4A-AS1 regulates glycolysis and glutaminolysis by mediating nucleolar translocation of HDAC2. EBioMedicine. 41:200–213. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Yoshida GJ: Emerging roles of Myc in stem cell biology and novel tumor therapies. J Exp Clin Cancer Res. 37:1732018. View Article : Google Scholar : PubMed/NCBI

95 

Dang CV: Gene regulation: Fine-tuned amplification in cells. Nature. 511:417–418. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Sabò A, Kress TR, Pelizzola M, de Pretis S, Gorski MM, Tesi A, Morelli MJ, Bora P, Doni M, Verrecchia A, et al: Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature. 511:488–492. 2014. View Article : Google Scholar

97 

Walz S, Lorenzin F, Morton J, Wiese KE, von Eyss B, Herold S, Rycak L, Dumay-Odelot H, Karim S, Bartkuhn M, et al: Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature. 511:483–487. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Dang CV, Kim JW, Gao P and Yustein J: The interplay between MYC and HIF in cancer. Nat Rev Cancer. 8:51–56. 2008. View Article : Google Scholar : PubMed/NCBI

99 

Grüning NM, Lehrach H and Ralser M: Regulatory crosstalk of the metabolic network. Trends Biochem Sci. 35:220–227. 2010. View Article : Google Scholar

100 

Kim JW, Gao P, Liu YC, Semenza GL and Dang CV: Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol. 27:7381–7393. 2007. View Article : Google Scholar

101 

Xiao ZD, Han L, Lee H, Zhuang L, Zhang Y, Baddour J, Nagrath D, Wood CG, Gu J, Wu X, et al: Energy stress-induced lncRNA FILNC1 represses c-Myc-mediated energy metabolism and inhibits renal tumor development. Nat Commun. 8:7832017. View Article : Google Scholar : PubMed/NCBI

102 

Liao B, Hu Y and Brewer G: Competitive binding of AUF1 and TIAR to MYC mRNA controls its translation. Nat Struct Mol Biol. 14:511–518. 2007. View Article : Google Scholar

103 

Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, et al: Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 20:285–295. 2018. View Article : Google Scholar

104 

Guo C, Shi H, Shang Y, Zhang Y, Cui J and Yu H: LncRNA LINC00261 overexpression suppresses the growth and metastasis of lung cancer via regulating miR-1269a/FOXO1 axis. Cancer Cell Int. 20:2752020. View Article : Google Scholar

105 

Yu Y, Li L, Zheng Z, Chen S, Chen E and Hu Y: Long non-coding RNA linc00261 suppresses gastric cancer progression via promoting Slug degradation. J Cell Mol Med. 21:955–967. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Yan D, Liu W, Liu Y and Luo M: LINC00261 suppresses human colon cancer progression via sponging miR-324-3p and inactivating the Wnt/β-catenin pathway. J Cell Physiol. 234:22648–22656. 2019. View Article : Google Scholar

107 

Zhai S, Xu Z, Xie J, Zhang J, Wang X, Peng C, Li H, Chen H, Shen B and Deng X: Epigenetic silencing of LncRNA LINC00261 promotes c-myc-mediated aerobic glycolysis by regulating miR-222-3p/HIPK2/ERK axis and sequestering IGF2BP1. Oncogene. 40:277–291. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Wang Y, Lu JH, Wu QN, Jin Y, Wang DS, Chen YX, Liu J, Luo XJ, Meng Q, Pu HY, et al: LncRNA LINRIS stabilizes IGF2BP2 and promotes the aerobic glycolysis in colorectal cancer. Mol Cancer. 18:1742019. View Article : Google Scholar : PubMed/NCBI

109 

Zhou Y, Li Y, Wang N, Li X, Zheng J and Ge L: UPF1 inhibits the hepatocellular carcinoma progression by targeting long non-coding RNA UCA1. Sci Rep. 9:66522019. View Article : Google Scholar : PubMed/NCBI

110 

Koller-Eichhorn R, Marquardt T, Gail R, Wittinghofer A, Kostrewa D, Kutay U and Kambach C: Human OLA1 defines an ATPase subfamily in the Obg family of GTP-binding proteins. J Biol Chem. 282:19928–19937. 2007. View Article : Google Scholar : PubMed/NCBI

111 

Whitaker-Menezes D, Martinez-Outschoorn UE, Flomenberg N, Birbe RC, Witkiewicz AK, Howell A, Pavlides S, Tsirigos A, Ertel A, Pestell RG, et al: Hyperactivation of oxidative mitochondrial metabolism in epithelial cancer cells in situ: Visualizing the therapeutic effects of metformin in tumor tissue. Cell Cycle. 10:4047–4064. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2022
Volume 48 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu W and Wu W: Research progress on the interaction between long non‑coding RNAs and RNA‑binding proteins to influence the reprogramming of tumor glucose metabolism (Review). Oncol Rep 48: 153, 2022
APA
Wu, W., & Wu, W. (2022). Research progress on the interaction between long non‑coding RNAs and RNA‑binding proteins to influence the reprogramming of tumor glucose metabolism (Review). Oncology Reports, 48, 153. https://doi.org/10.3892/or.2022.8365
MLA
Wu, W., Wen, K."Research progress on the interaction between long non‑coding RNAs and RNA‑binding proteins to influence the reprogramming of tumor glucose metabolism (Review)". Oncology Reports 48.3 (2022): 153.
Chicago
Wu, W., Wen, K."Research progress on the interaction between long non‑coding RNAs and RNA‑binding proteins to influence the reprogramming of tumor glucose metabolism (Review)". Oncology Reports 48, no. 3 (2022): 153. https://doi.org/10.3892/or.2022.8365