Skip to main content

The roles and molecular mechanisms of non-coding RNA in cancer metabolic reprogramming

Abstract

One of the key features of cancer is energy metabolic reprogramming which is tightly related to cancer proliferation, invasion, metastasis, and chemotherapy resistance. NcRNAs are a class of RNAs having no protein-coding potential and mainly include microRNAs, lncRNAs and circRNAs. Accumulated evidence has suggested that ncRNAs play an essential role in regulating cancer metabolic reprogramming, and the altered metabolic networks mediated by ncRNAs primarily drive carcinogenesis by regulating the expression of metabolic enzymes and transporter proteins. Importantly, accumulated research has revealed that dysregulated ncRNAs mediate metabolic reprogramming contributing to the generation of therapeutic tolerance. Elucidating the molecular mechanism of ncRNAs in cancer metabolic reprogramming can provide promising metabolism-related therapeutic targets for treatment as well as overcome therapeutic tolerance. In conclusion, this review updates the latest molecular mechanisms of ncRNAs related to cancer metabolic reprogramming.

Introduction

Cancer is one of the most common fatal diseases, which seriously threatens human lives and health. According to the most recent data in 2023, about two million new cancer cases and almost sixty thousand cancer deaths are projected to occur in the United States [1]. Thus, it is urgent to find new biomarkers and therapeutic strategies for cancer with great significance.

Currently, energy metabolic reprogramming, one of the Hallmarks of cancer, has become the hot spot in the research field [2]. In 1921, German scientists discovered the “Warburg effect”, also known as “aerobic glycolysis”, which meant that apart from normal cellular oxidative phosphorylation, cancer cells undergo aerobic glycolysis under aerobic conditions by taking in massive glucose to provide numerous Adenosine triphosphate (ATP) and biosynthetic materials for rapid proliferation. It is a huge breakthrough in the field of cancer metabolism research. Besides glucose metabolism reprogramming, cancer cells can also induce the alterations of lipids and amino acids et al. to meet the requirements for malignant progress. The altered energy metabolism of cancer cells can reshape the cancer microenvironment, which facilitates the maintenance of malignant phenotype and immune escape [3, 4]. Importantly, targeting specific metabolic pathways of cancer cells or simultaneously synergizing other therapeutic strategies is one of the effective means to improve the efficacy of cancer therapy [5, 6].

Non-coding RNAs (ncRNAs) are novel gene regulatory molecules, mainly including miRNAs, long non-coding RNAs (lncRNAs) and circular non-coding RNA (circRNAs) et al. In recent years available evidence is increasingly indicating that dysregulated ncRNAs play a significant role in the progression of cancer malignancy through metabolic reprogramming [7, 8]. Consequently, there is a pressing need to elucidate the potential regulatory mechanisms of ncRNAs in the cancer metabolic network for the development of carcinogenesis, as this knowledge holds great promise for future prevention and treatment strategies. This review aims to provide an updated overview of the latest research progress on the molecular mechanisms of ncRNAs in cancer energy metabolic reprogramming.

Overview of metabolic reprogramming in cancer cells

It is known that energy metabolism undergoes great changes during the development of cancer, referring to the “Warburg effect”, the production of vast amounts of ATP and lactate by aerobic glycolysis of cancer cells under aerobic conditions through massive glucose uptake to support the acquisition and maintenance the malignant phenotype [9]. The initiation of aerobic glycolysis is facilitated by the glucose transporter1 (GLUT1) mediated abnormal transportation of glucose and the abnormal expression of enzymes involved in glycolysis. This phenomenon is induced by oncogenes like c-Myc and HIF-1α, whereas cancer suppressors such as p53 and OVOL2 exhibit an opposing effect. The activation of aerobic glycolysis results in the provision of substantial resources for cancer cells, encompassing ATP and metabolic intermediates derived [10,11,12,13,14,15]. In addition to facilitating the malignant phenotype of cancer cells, lactate, a byproduct of aerobic glycolysis in cancer cells, can be extracellularly transported through monocarboxylate transporter protein 4. This transport mechanism contributes to the creation of an acidic microenvironment within cancer cells, ultimately leading to the polarization of M2 macrophages and the promotion of immunosuppressive function in Treg cells [3]; Conversely, it inhibits the activation of cytotoxic T cells and depletes the interferon-γ (IFN-γ) secretion function of natural killer (NK) cells [3, 16, 17]. Furthermore, increased lactate abundance in the tumor microenvironment (TME) also promotes cancer immune escape through epigenetics [18]. In conclusion, glucose metabolic reprogramming promotes the malignant progression of cancer cells and its targeting can be a promising strategy to suppress cancer progression.

The mechanisms of lipids including absorption, synthesis, and oxidation in cancer cells primarily distinct from normal cells [19]. These processes are extensively regulated by transcription factors such as peroxisome proliferator-activated receptors and sterol regulatory progenitor proteins [20, 21]. Cancer cells require a large amount of lipids and cholesterol to meet their raised demand by increasing the uptake of exogenous lipids, and lipoproteins and activating the expression of enzymes related to the de novo synthesis of lipids and cholesterol. The increased expression of fatty acids in vivo can modify proteins at the epistatic level. An emerging study shows that palmitoylation modification of GLUT1 regulates its plasma membrane localization impairing glycolysis and cell proliferation in cancer cells [22]. Furthermore, ferroptosis is driven by the peroxidation of polyunsaturated fatty acids catalyzed by ferrous ions [23]. Changes in lipid metabolism in cancer cells promote aberrant accumulation of lipids in TME, which in turn promotes unusual accumulation of lipids in cancer-infiltrating immune cells leading to the translation between immunosuppressive and anti-inflammatory phenotype [5, 24,25,26]. Intriguingly, the abnormal accumulation of cholesterol in cancer cells can contribute to the stable expression of programmed death ligand-1 (PD-L1) by binding to cholesterol-recognition amino acid consensus motifs on PD-L1, resulting in the development of cancer immune escape [5]. As its low response to immunotherapy, targeting lipid metabolism in cancer may be a significant approach to improve the efficiency of immune therapy and inhibit cancer progression.

The metabolic reprogramming of amino acids plays an essential role in the development of cancer. In addition, amino acids affect the function of immune cells in anti-cancer immunity [27]. Accumulated evidence shows that most cancer cells strongly depend on glutamine known as “glutamine addiction” as it stagnates in growth and even dies once cancer cells are deprived of glutamine [28]. Cancer cells reduce the activation and infiltration of CD8+ T cells and recruit myeloid-derived suppressor cells (MDSCs) by increasing the secretion of granulocyte colony-stimulating factor through the massive consumption of glutamine in TME [3]. However, restricting glutamine uptake by cancer cells increases PD-L1 expression through stimulation of Ca2+/NF-κB signaling, resulting in diminished anticancer activity of T cells, which may be successfully eliminated by anti-PD-L1 therapy [6]. The altered metabolism of serine, tryptophan and glycine in cancer cells has likewise received considerable attention. The classically activated macrophages (M1) polarization of macrophages induced by IFN-γ can be significantly enhanced by inhibiting the ab initio synthesis of serine or by limiting the uptake of exogenous serine [29]. Serine is the main substrate of one-carbon units for nucleotide synthesis in vivo, however, in cancer cells with high expression of indoleamine 2, 3-dioxygenase 1(IDO1), tryptophan could replace serine as the main substrate of one-carbon units [30]. High levels of IDO and tryptophan 2,3-dioxygenase (TDO) in cancer inhibit the cancer-killing effects of T cells by reducing tryptophan levels in TME [31]. Related studies have proved that immunotherapy induces IFN-γ increasing release by activation of CD8+ T cells, downregulates the expression of the xc-subunit of the glutamate-cystine reverse transport system and weakens the uptake of cystine by cancer cells, thereby promoting cancer cell death [32]. Therefore, it may have the potential to inhibit the malignant progression of cancer cells as well as improve the efficacy of anticancer therapy by targeting specific amino acid metabolic pathways.

In addition, aberrant activation of oncogenes can promote anabolism through multiple signaling pathways, such as PI3K/Akt/mTOR, HIF-1α and sterol-regulatory element binding proteins (SREBP) et al. [33]. TME provides the fundamental materials for cancer growth, and cancer metabolic reprogramming changes the level of substances in TME, indirectly affecting the release of inflammatory factors and anti-cancer function of immune cells in TME [34]. In conclusion, as metabolic reprogramming of cancer cells facilitates the development of metabolic phenotype for malignant progression. it may be a potential strategy to effectively suppress their malignant progression by changing the metabolic phenotype of cancer.

Overview of NcRNAs

For decades, ncRNAs, used to be regarded as by-products of transcription, have been found to play essential roles in a variety of important cellular life activities, which include chromatin remodeling, transcription, post-transcriptional modifications (PTMs) and signal transduction [35,36,37,38,39]. NcRNAs refer to a class of RNAs that have no protein-coding functions in cells, including ribosomal RNA (rRNA), transfer RNA (tRNA), small nuclear RNA (snRNA), and small nucleolar RNA (snoRNA), piRNA (PIWI-interacting RNA), microRNA (miRNA), lncRNAs, and circ RNAs [40, 41]. In this review, we mainly focus on microRNAs, lncRNAs and circ RNAs.

MicroRNAs are a set of ncRNAs with 20nt nucleotide and play a key regulatory role at the post-transcriptional level of messenger RNA (mRNA) by binding to the 3’-untranslated regions (3’-UTR) of the target gene to degrade the mRNA of the target gene or inhibit the translation process of the target gene [42, 43]. LncRNAs are a class of special RNA molecules that are longer than 200 nucleotides and without protein-coding potential, and their biological generation is similar to mRNAs [44]. However, due to the splicing efficiency of lncRNAs being lower than mRNAs, most lncRNAs are retained in the nucleus after transcription involved in mediating interactions between DNA and proteins, adsorbing microRNAs, and binding to proteins as decoys, which contribute to regulating chromatin structure, remodeling chromatin and chromatin modification. And only a small fraction is exported to the cytoplasm [45,46,47]. CircRNAs are novel ring-like molecules formed by reverse shearing of exons. The 3’ and 5’ ends of circ RNA are linked to form a covalently closed loop structure, which confers a longer half-life, more stability and less sensitivity to RNA exonucleases. Similar to lncRNAs, circRNAs have played an important regulatory role in gene expression at epigenetic, transcriptional or post-transcriptional levels [48,49,50]. There is sufficient evidence showing that ncRNAs are key regulators of metabolic reprogramming, which results in cancer rapid development [51,52,53]. Importantly, some ncRNAs serve as metabolic-related oncogenes, while some other ncRNAs act as metabolic-related suppressors. Thus, it is urgent to figure out the potential molecule mechanism of dysregulated metabolism mediated by ncRNAs in carcinogenesis, which may be a promising strategy for therapy in the future.

NcRNAs affect cancer progression through metabolic reprogramming

Numerous research has demonstrated that dysregulated ncRNAs related to metabolic networks can promote or suppress carcinogenesis, by facilitating cancer to acquire and maintain aggressive malignant phenotype and to target multiple metabolic signaling pathways [54,55,56,57,58,59,60,61,62]. Therefore, we update the latest research progress on the mechanisms of ncRNAs in metabolic reprogramming from three perspectives: glucose, lipid and amino acids, respectively.

The effects of ncRNAs on cancer glucose metabolic reprogramming

It has been well-documented that abnormal glucose metabolism is one of the leading causes of cancer development [63]. Usually, normal tissues acquire energy through aerobic oxidative phosphorylation but undergo anaerobic glycolysis under hypoxia. However, the main way to acquire energy in cancer cells is aerobic glycolysis even under the condition of adequate oxygen. Accumulated evidence suggests there are numerous glucose metabolism-associated ncRNAs dysregulated in cancers resulting in mitochondrial dysfunction, abundant activation of key enzymes, altered isozyme profiles and dysregulated glucose metabolic signalling pathways [64,65,66]. Current progress in the role of ncRNAs in glucose metabolism has been summarized in Fig. 1.

Fig. 1
figure 1

Dysregulated ncRNAs involve in glucose metabolism

NcRNAs and glucose uptake

GLUT is a key transporter protein for glucose uptake. Extensive research has demonstrated that dysregulation of ncRNAs in cancer mediates aberrant expression of GLUT1, such as miR-181a and let-7a-5p. The increased expression level of miR-181a promotes metastasis in CRC by suppressing phosphatase and tensin homolog (PTEN) expression and promoting GLUT1 expression [67]. In contrast, the low expression level of let-7a-5p in triple-negative breast cancer (TNBC) patients was negatively correlated with GLUT12 and promoted the proliferate, migrate, and invade ability of TNBC cells [68]. It is suggested that circRNF13 plays an essential role in the glucose metabolism programming of nasopharyngeal carcinoma (NPC) by stabling SUMO2 mRNA, increasing the SUMO and ubiquitination degradation of GLUT1 [69]. Li et al. found that increased expression of lncRNA GAL promotes colorectal cancer (CRC) liver metastasis. Mechanically, GAL interacts with GLUT1 to increase the SUMOylation of GLUT1 and thereby promote the stability of its mRNA [70]. Moreover, studies have revealed that circDENND4C promotes GLUT1 expression by sponging miR-760 contributing to the rapid proliferation of CRC [71]. Cui and his colleagues have illustrated that m6A modification of circFOXK2 promotes stability of GLUT1, which in turn plays a carcinogenic role in oral squamous cell carcinoma [72]. Above all, dysregulated ncRNAs affect GLUT-mediated glucose uptake rate in cancer cells related to malignant progression.

NcRNAs and glycolysis

Hexokinase (HK) is the first rate-limiting enzyme in glycolysis. The increased expression of HK2 in cancer is positively correlated with cancer malignancy, which is regulated by numerous ncRNAs. A recent study has revealed that increased expression of lncRNA KCNQ1OT1 in CRC promotes proliferation by decreasing the ubiquitination level of HK2, resulting in increased aerobic glycolysis [73]. LncRNA MIR17HG promotes liver metastasis through miR-138-5p/HK1 axis, which induces glycolysis. Intriguingly, the product of glycolysis, lactate, can facilitate the transcription of MIR17HG by activating the p38/Elk-1 signaling pathway, forming a positive feedback loop [74]. Another study related to lung adenocarcinoma has suggested that upregulation of lncRNA FAM83A-AS1 promotes the expression of HK2 through sponging miR-202-3p, leading to increased glycolysis [75]. A similar effect of circ0000231 on aerobic glycolysis in CRC cells has been illustrated in the research by Liu et al. [76]. According to the latest findings above, by targeting the lncRNA-miRNA-mRNA competitive endogenous RNA (ceRNA) regulatory network, which modulates HK expression could be a promising strategy to inhibit cancer malignant progression.

Phosphofructokinase-1 is one of the key enzymes in the glycolytic process. Cui et al. has reported that upregulated expression of lncRNA MAFG-AS1 in CRC promotes cancer proliferation, cell cycle progression, and inhibits cell apoptosis by targeting the miR-147b/NDUFA4 axis to upregulate phosphofructokinase-1 (PFK-1) expression [77]. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), encoded by the pfkfb3 gene, is abundantly expressed and closely related with the generation of chemotherapy resistance. There is sufficient evidence showing that dysregulated ncRNAs are involved in regulating the expression of PFKFB3. Deng et al. have demonstrated that increased expression of miR-488 in CRC can inhibit glucose uptake and lactate secretion by targeting PFKFB3 and improve CRC sensitivity to oxaliplatin/5-Fu [78]. In addition, the latest identified circSAMD4A and circ0014130, which are highly expressed in CRC, contribute to the 5-Fluorouracil (5-Fu) resistance by acting as ceRNAs resulting in the increased expression of PFKBF3 [79, 80]. In conclusion, there is sufficient evidence showing that PFKBF3 dysregulation mediated by ncRNAs is closely associated with chemoresistance in cancers. Importantly, targeting ncRNAs may be an effective strategy to overcome chemotherapy resistance caused by high expression of PFKFB3.

Recent studies have reported that ncRNAs generated from intestinal microbial are involved in cancer development [81, 82]. F.nucleatum promotes the transcription factor SP1 to bind with the promoter region of lncRNA ENO1-IT1 to activate ENO1-IT1 transcription causing increased glucose metabolism in CRC to further support carcinogenesis. It suggests a complicated interaction between ncRNAs deriving from microbiome and glucose metabolism [83]. Besides, alpha-enolase (ENO1) has been identified by Ma et al. acts as the target of lncRNA ANRIL/miR-125a pathway inducing drug resistance in TNBC cells [84]. Wang et al. found that mesenchymal stem cells (MSCs) promoted cancer progression by transporting exosomes containing LINC01559 to gastric cancer cells. Increased expression of LINC01559 activates the PIK3/AKT pathway by sponging miR-1343-3p to upregulate phosphoglycerate kinase 1 (PGK1). In addition, LINC01559 induces methylation of the PTEN promoter, which triggers the PIK3/AKT signaling pathway by inhibiting PTEN and accelerates cancer progression [85].

Pyruvate kinase is another key enzyme in glycolysis. Recent research has reported that miR-206 and lncRNA SNHG6 play oncogenic and pro-cancer roles in CRC by targeting heterogeneous ribonucleic acid proteins to alter pyruvate kinase isozyme gene splicing patterns and control the PKM1/PKM2 ratio, respectively [86, 87]. The small peptide HOXB-A3 encoded by lncRNA HOXB-AS3 is revealed to inhibit the splicing formation of pyruvate kinase M2 (PKM2) by competitively binding with the arginine residue in the hnRNP A1 RGG motif to exert cancer suppressive effects [88]. LncRNA FEZF1-AS1 enhances the stability of PKM2 by binding to its A2 structural domain, leading to elevated PKM2 levels in the cytoplasm and nucleus. FEZF1-AS1 induces increased PKM2 in the cytoplasm to promote aerobic glycolysis, and in the nucleus to further activate the signal transducer and activator of transcription 3 (STAT3) signaling pathway [89]. Hua et al. have shown that upregulation of lncRNA AC020978 is transcriptionally activated by HIF-1α in non-small cell lung cancer, which interacts with PKM2 and enhances the stability of PKM2 protein. In addition, AC020978 can also increase the interaction of PKM2 with HIF-1α by promoting the nuclear translocation of PKM2, further enhancing the transcriptional activation of HIF-1α on target genes [90]. NcRNAs regulate the Warburg effect by alternatively splicing pyruvate kinase isozymes and controlling the ratio of PKM1/PKM2. Targeting ncRNAs to regulate the splicing of PKM may be an effective strategy to suppress aerobic glycolysis.

Apart from the above-mentioned key enzymes and transporters in glycolysis, lactic dehydrogenase is a critical rate-limiting enzyme for glycolysis. It is identified that lncRNA MCF2L-AS1 promotes the expression of heat shock protein 90 (HSP90) by sponging miR-874-3p in CRC cells leading to the increased production of lactic acid [91]. Tang et al. found that lncRNA GLCC1 reduces ubiquitination of c-Myc by directly interacting with HSP90 to modify the transcriptional pattern of the target gene lactate dehydrogenase A (LDHA), thereby reprogramming glucose metabolism to promote CRC progression [92]. Interestingly, it is reported that a c-Myc-regulated lncRNA glycoLINC can act as a backbone to assemble several glycolysis-related enzymes into a metabolon that exerts a pro-carcinogenic effect by increasing the catalytic efficiency of glycolytic enzymes leading to an increase in glycolytic flux [93]. The increased expression of LDHA is regulated by many oncogenes, promoting glycolysis and malignant progression of cancers, which is mediated by ncRNAs. Thus, targeting dysregulated ncRNAs may be potential targets to suppress cancer development through regulating the expression of LDHA.

NcRNAs and the pentose phosphate pathway

The PPP is an important metabolic bypass for glucose and glucose-6-phosphate dehydrogenase (G6PD) is the key rate-limiting enzyme. A recently published study has suggested that LINC01615 in CRC increased pentose phosphate pathway (PPP) flux, reduced reactive oxygen species (ROS) production, and promoted nucleotide and lipid synthesis by regulating the splicing of G6PD precursor mRNA [94]. Chen et al. discovered that circ0003215 suppressed PPP and the malignant phenotype of CRC through the miR-663b/ DLG4 axis. Mechanistically, DLG4, which is mediated by circ0003215 and upregulated by K48-linked G6PD, increased its ubiquitinated degradation and inhibited the PPP [95]. In osteosarcoma, lncRNA OR3A4 increases G6PD expression through sponging miR-1207-5P, leading to the activation of PPP, thus promoting cancer cell growth [96]. In addition, the short peptide pep-AP encoded by lncRNA AP has been validated to facilitate oxaliplatin by interacting with TALDO1, decreasing NADPH/NADP+ and glutathione (GSH) levels [97]. Yang et al. revealed that high expression of lncRNA PDIA3P in multiple myeloma binds to c-Myc to increase its transcriptional activity and promotes c-Myc interacting with the G6PD promoter, leading to increased G6PD expression and malignant progression [98]. In a word, ncRNAs play a critical role in sustaining nucleotide biosynthesis as well as maintaining intracellular redox homeostasis through the PPP pathway. However, the PPP pathway mediated by dysregulated ncRNAs can provide numerous synthetic raw materials for cancer proliferation, while blocking this can be a promising strategy for cancer treatment.

NcRNAs and the tricarboxylic acid cycle

Mitochondria are vital for the complete oxidative phosphorylation of glucose. After glucose is converted to pyruvate via the glycolytic route, the pyruvate dehydrogenase complex (PDC) in the mitochondria catalyzes the formation of acetyl CoA, which enters the tricarboxylic acid cycle (TAC). In malignancies, abnormal activation of pyruvate dehydrogenase kinases (PDKs), inhibitors of pyruvate dehydrogenase, leads to increased glycolytic flow. PDC activity is inhibited in cancer cells by phosphorylation of the E1a subunit of PDKs, restricting pyruvate entrance into mitochondria [99].

In CRC, an lncRNA HCG11/ miR-144-3p/ PDK4 axis has been illustrated to promote aerobic glycolysis and lactate generation [100]. LncRNA SPRY4-IT1 is documented to bind to PDK1 and boost its stable expression, resulting in enhanced aerobic glycolysis and accelerated CRC progression [101]. Succinate dehydrogenase (SDH) in the TAC is regulated by ncRNA such as miR-142-5p, which is increased in CRC and diminishes oxygen intake while increasing lactate generation by targeting SDHB [102]. Let-7a enriched in extracellular vesicles (EVs) hampers oxidative phosphorylation of mitochondria and CRC development via the Lin28a/SDHA signaling pathway [103]. Dysregulation of enzymes such as PDK and SDH mediated by ncRNAs in cancers regulate aerobic glycolysis by limiting pyruvate access to mitochondria for complete oxidative phosphorylation. It may play a key role in cancer treatment in the future.

Defective mitochondrial function and abnormal biogenesis are often also important factors in promoting aerobic glycolysis, which is regulated by nuclear respiratory factors 1 and 2 (NRF1 and NRF2), mitochondrial transcription factor A (TFAM) and ncRNAs [104]. According to a recently released study, LINC00467 encodes an ATP synthase-associated peptide (ASAP), which interacts with the ATP5A and ATP5C to increase ATP synthase activity and mitochondrial oxygen consumption rate [105]. Lnc 00839 functions as a scaffold in the nucleus, enhancing the transcriptional activity of NRF1, which has a pro-carcinogenic impact on CRC by promoting oxidative phosphorylation (OXPHOS) and mitochondrial formation [47]. Reduced ROS and NAD+/NADH have been reported as a result of miR-483, miR-676, and miR-877 downregulating the expression of mitochondrial metabolism-related genes [106]. According to Zhao et al., the nuclear-encoded lncRNA MALAT1, aberrantly enriched in hepatocellular carcinoma cells, promotes the synthesis of mitochondrial DNA (mtDNA) by binding to it and altering its methylation and mitochondrial function, thus controlling mitochondrial metabolism at the epigenetic level [107]. Given all this, targeting abnormal mitochondria through ncRNAs offers great possibilities for treating cancers.

The effects of ncRNAs on cancer lipid metabolic reprogramming

Accumulated studies have demonstrated that dysregulated ncRNAs are involved in the progression of cancer lipid metabolic reprogramming including abnormal lipid uptake, accumulation, synthesis and oxidation et.al. [108,109,110,111,112]. The role of ncRNAs in lipid metabolism has been summarized in Fig. 2.

Fig. 2
figure 2

Dysregulated ncRNAs involve in lipid metabolism

NcRNAs and lipid uptake and accumulation

The uptake of exogenous fatty acids is mainly dependent on the transmembrane transport of CD36, fatty acid transport proteins (FATPs) and fatty acid binding proteins (FABPs). In contrast, cellular uptake and utilization of cholesterol are dependent on the low-density lipoprotein receptor superfamily (LDLRs). It has been reported that miR-107 overexpressed in CRC cells decreases CD36 expression by inhibiting the PPAR signaling pathway. In contrast, lncRNA TINCR, an endogenous competing RNA for miR-107, inhibits its expression and activates CD36 to play a role in inhibiting cell proliferation and promoting apoptosis [110].

The decreased expression of very low-density-lipoprotein receptor (VLDLR) in cancer cell membranes contributes to the accumulation of cholesterol in the cytoplasm. It has been found that VLDLR downregulation promotes CRC cell proliferation and migration through elevated miR-200c expression [113]. Mosapour et al. found that miR-4465 targets VLDLR to promote the development of breast cancer, resulting in cholesterol accumulated in cytoplasm [114]. Abnormally high cholesterol in hepatocellular carcinoma cells promotes malignant lesions in non-alcoholic fatty liver disease (NAFLD) by inducing lncRNA SNHG6 to localize in the endoplasmic reticulum-lysosome interactions region and activating the mammalian target of rapamycin (mTORC1) signaling pathway [115]. In contrast, LncRNA Mexis and LINC DYNLRB2-2 induce macrophage ABCA1 gene expression, which in turn promotes ABCA1-mediated cholesterol efflux leading to increased cholesterol levels in TME, which may further promote cholesterol uptake by cancer cells [116, 117]. Above all, high lipid accumulation in cancer cells promotes the development of cancer. Therefore, limiting lipid uptake and abnormal intracellular lipid accumulation through ncRNAs may be a promising strategy to improve cancer treatment efficiency.

NcRNAs and lipid synthesis

It has been shown that lncRNA SNHG16 targets various lipid metabolizing enzymes such as stearoyl-CoA desaturase (SCD) and ATP-citrate synthase (ACLY) through endogenous competing RNAs in CRC. And high expression of SNHG16 positively correlates with the expression of transcription factors long-chain-fatty-acid-CoA ligase 2 (ASCL2), ETS2 and c-Myc, which are activated by the Wnt signaling pathway [111]. Acetyl coenzyme A carboxylase (ACC), a rate-limiting enzyme for fatty acid synthesis, has been found transcriptional activation by circCAPRIN1 in CRC through direct binding to STAT2, causing increased lipid synthesis and CRC progression [118]. Peng et al. demonstrated that LncRNA FASR expression was driven by USF1 via a super-enhancer. FASRL enhances fatty acid synthesis and lipid accumulation by binding to the fatty acid synthesis rate-limiting enzyme acetyl coenzyme A carboxylase (ACAC), thereby promoting hepatocellular carcinoma (HCC) [119]. A study has revealed that miR-BART18-3p, encoded by EBV, activates the HIF-1α/LDHA axis by targeting sirtuin to promote lactate accumulation and acetyl coenzyme A production under hypoxic conditions, resulting in enhanced acetylation of histones H3K9, H3K14 and H3K27 of fatty acid synthase (FASN) and activates de novo synthesis of fat [120]. Accumulated research has proved that dysregulated ncRNAs play an indispensable role in regulating lipid metabolism.

The mevalonate pathway is a key signaling pathway for cholesterol synthesis, which can not only be activated by MYC but also lead to up-regulation of MYC expression through up-regulation of miR-33b, forming a positive feedback loop [121]. MiR-195 has been illustrated that effectively prevent the accumulation of cholesterol by inhibiting the expression of IKKα and TAB3 [122]. A previous study has revealed that lncRNA 030 maintains the stemness of breast cancer stem cells by interacting with PCBP2, regulating the stability of squalene monooxygenase (SQLE) mRNA, promoting cellular cholesterol synthesis, and further leading to activation of the PI3K/AKT pathway [123]. Ni et al. revealed that an HMG-CoA reductase (HMGCR) reductase inhibitor, simvastatin, could promote anticancer effects by inhibiting lncRNA SNHG29-mediated Yes-Associated Protein (YAP) activation and suppressing PD-L1 expression [124]. It has been demonstrated that excessive accumulation of cholesterol induces PD-L1 expression in cancer cells [5]. Therefore, inhibition of cancer cell cholesterol synthesis pathway by targeting ncRNAs may be an emerging direction to improve the efficacy of immunotherapy. Furthermore, Wang et al. found that lncRNA ZFAS1 increased the expression level of SCD1 and FASN, through bound PABP2 to stabilize the mRNA stability of SREBP1, leading to promoting lipid accumulation in CRC [125]. Highly expressed RPRD1B in gastric cancer cells increases fatty acid uptake and synthesis through transcriptional upregulation of c-Jun/c-Fos and activation of the c-Jun/c-Fos/SREBP1 axis [126]. Liu et al. have shown that upregulated circPRKAA1 can form a tetrameric complex with Ku80/Ku70 heterodimer and mSREBP-1 increasing the de novo synthesis of lipids through transcriptional activation of ACC1, ACLY, SCD1 and FASN [127]. Generally, increased lipid synthesis is tightly related to cancer progression, which can be induced by dysregulated ncRNAs through targeting lipid-related enzymes and their key components SREBPs.

NcRNAs and lipid oxidation

It has been demonstrated by Gharib et al. that miR-497-5p plays an oncogenic role in CRC by targeting ACSL5 to block the oxidation activation of fatty acids [112]. Barisciano et al. research suggests that miR-27a may act by targeting carnitine palmitoyltransferase-1 (CPT-1) and acyl coenzyme a dehydrogenase-9 (ACAD9) in the process of fatty acid β-oxidation. PPARγ coactivator-1α (PGC-1α), which directly regulates peroxisome proliferators-activated receptors (PPARγ), is increased in CRC cells when miR-27a is knocked down. Also, protein levels of the downstream effectors of PPARγ, CPT1A and ACAD9, are upregulated [128]. Li et al. showed that in the condition of serum deprivation, highly expressed circRNAACC1 acted as a cancer-promoting factor to control fatty acid β oxidation of CRC by activating AMP-activated protein kinase (AMPK) [129].

Ferroptosis, which develops from an increase in iron-dependent lipid peroxide, is regulated by a network of ncRNAs [130, 131]. According to Luo et al., LINC01606 serves as the ceRNA of miR-423-5p to upregulate SCD1 and further stimulate the Wnt/β-catenin signaling pathway, which prevents ferroptosis in CRC cells and preserves cell stemness [132]. Zhang et al. revealed that the ferroptosis inducers Erastin and RSL3 promoted lncRNA NEAT1 expression by facilitating p53 binding to its promoter, increasing intracellular ROS production [133]. It has been reported that LINC00239 resists ferroptosis by interacting with the Kelch structural domain of Keap1, thereby enhancing Nrf2 protein stability promoting its nuclear translocation and further reducing the activity of Erastin and RSL3 [134]. Zhang et al. reported that curcuminol can induce ferroptosis in lung cancer cells through lncRNA H19/miR-19b-3p/FTH1 axis [135]. Collectively, cell death plays a significant role in several biological processes and the targeting of cell death emerges as a prominent therapeutic strategy for the treatment of a variety of diseases, especially cancers [131]. Ferroptosis, an emerging type of cell death, is distinct from apoptosis and necrosis in the conventional sense [131]. Thus, revealing the molecular mechanisms of lncRNAs mediated-ferroptosis could provide new insights into the cell death processes and potential molecular components to develop promising strategies to prevent and stimulate cell death.

The effects of ncRNAs on amino acid metabolic reprogramming

Emerging studies have revealed that altered amino acid metabolism is closely associated with cancer outgrowth, metastasis, and therapeutic resistance [136, 137]. On another hand, numerous studies have shown that dysregulated ncRNAs are involved in regulating the amino acid metabolic reprogramming, which facilitates cancer progression [138, 139]. The latest progress in the role of ncRNAs in amino acid metabolism has been summarized in Fig. 3.

Fig. 3
figure 3

Dysregulated ncRNAs involve in amino acid metabolism

NcRNAs and glutamine

Rapidly proliferating cancer cells often exhibit an extremely strong dependence on glutamine [140]. It is Dong et al. show that miR-137 directly regulates amino-acid transporter 2 (ASCT2) expression, which significantly reduces CRC cells glutamine uptake [141].

Glutaminase (GLS) is an amino acid enzyme that has been identified as a target of miR-137. Heat shock factor 1 (HSF1) can inhibit the interaction of miR-137 and GLS to promote glutamine catabolism and activate mTOR signaling, which contributes to cancer development [138]. Liu et al. have reported that lncRNA HOX-AS1 can increase the expression of GLS and the levels of intracellular glutamine metabolism through sponging miR-126-5p [139]. Moreover, the latest research has demonstrated that lncRNA CAPRIN1 can inhibit GLS1 translation by driving CAPRIN1-mediated phase segregation in glutamine deficiency, preventing excessive depletion of limited glutamine to maintain cancer survival [142]. Wang et al. have demonstrated that downregulated miR-375 significantly promotes CRC development by activating the PI3K/Akt pathway and increasing the expression of glutamic–pyruvic transaminase (GPT2) [143, 144]. Limiting cancer cells to uptake glutamine can be an effective strategy to inhibit cancer growth, while ncRNAs play an important role in regulating glutamine metabolism. Therefore, targeting ncRNAs associated with glutamine metabolism is likely to be a potential approach to limit cancer progression.

NcRNAs and other amino acids

Apart from glutamine, there are numerous research revealed the potential role of ncRNAs in other amino acid metabolism.

Serine and glycine play an essential role in maintaining the anabolic requirements of cancer and supporting proliferation by controlling redox states [145]. Zhu et al. demonstrated that gLINC increased glycolytic flux under normal conditions and promoted cancer cell survival in serine deficiency [93]. Zhang G et al. has demonstrated that LncRNA 01564 is upregulated in HCC, which promotes the survival of HCC under glucose deprivation by targeting miR-107/103b-3p/ PHGDH axis [146]. According to Liu et.al, increased expression of circMYH9 binds the m6A reader hnRNPA2B1 into the nucleus, which reduces the stability of p53 pre-mRNA, while increasing endogenous serine production through enhancing phosphoglycerate dehydrogenase (PHGDH) activity. Amino acid deficiency leads to greater cellular production of ROS, which can enhance circMYH9 expression by attaching to the promoter region of circMYH9 and increasing endogenous serine production by increasing the expression of HIF-1α [147].

NcRNAs can also target metabolic pathways related to amino acids to exert regulatory effects. In esophageal squamous cell carcinoma, the increased LncRNA SLC25AS1-AS can not only maintain the stability of SLC25A21 mRNA but also reduce the intracellular NAD+/NADH ratio by affecting tryptophan catabolism [148]. Ornithine is the initiator of polyamine synthesis, increased expression of polyamine synthesis genes in cancer promotes cancer progression [149].

Ornithine decarboxylase 1 (ODC1), a crucial enzyme for polyamine biosynthesis using ornithine from the urea cycle, is frequently deregulated in cancer [150]. According to Meng et al. hypoxia-induced lncRNA LVBU accelerates CRC cell proliferation by regulating the BCL6-p53 axis and urea cycle/polyamine production pathway. LVBU overexpression can reduce the degradation of B-cell lymphoma 6 protein (BCL6) by sponging miR-10a/miR-34c, which deregulates p53 on arginase1, ornithine transaminase and ODC1 genes and further accelerates CRC cell proliferation [151].

According to a previous study, the high expression of LINC01234 encourages HCC proliferation and migration by interacting with the promoter of Argininosuccinate Synthase 1 (ASS1) and blocking p53-mediated transcriptional activation, which raises aspartate levels and activates the mTOR signaling pathway [152]. Li et al. has found that HBXIP protein interacts with c-Myc through the leucine zipper, recruiting lncRNA Hotair and histone demethylase LSD1 to transcriptionally activate c-Myc. Silencing Hotair blocked the growth-promoting effect of c-Myc in breast cancer cells [153]. Ma et al. has discovered that LINC01431 increases the interaction between PRMT1 and covalently closed loop DNA (cccDNA) of hepatitis B virus (HBV) to further limit HBVs replication by boosting H4R3me2a modification on cccRNA. The viral protein HBx maintains persistent HBV replication by escaping LINC01431-mediated transcriptional repression through the transcription factor ZHX2, which inhibits transcription of the host LINC01431 [154]. In addition, Yan et al. has shown that SRSP, encoded by lncRNA LOC90024, can bind with serine and arginine-rich splicing factor 3 (SRSF3) to regulate mRNA splicing [155]. In conclusion, amino acid metabolism regulated by ncRNAs has suggested novel targets for cancer therapy in the future. However, are more studies needed to figure out the potential mechanism of amino acid metabolic reprogramming mediated by dysregulated ncRNAs.

NcRNAs effect cancer therapeutic tolerance through metabolic reprogramming

Chemotherapy, radiation, targeted therapy and immunotherapy are the major strategies used for malignant cancers. However, the outcome of patients does not remain satisfactory due to limitations such as multi-drug resistance and tumor relapse [156]. Numerous types of research have implicated that ncRNAs may be a potential target to overcome therapeutic tolerance. Nevertheless, the potential regulatory mechanism for the occurrence of therapeutic tolerance and metabolic reprogramming in cancer needs to be further investigated.

NcRNAs mediate chemotherapy resistance through metabolic reprogramming

Accumulated studies have demonstrated that ncRNA-mediated metabolic reprogramming is involved in regulating the chemotherapy resistance and it has been summarized in Fig. 4 [7]. MiR-488 has been found to promote oxaliplatin and 5-Fu-induced CRC cell apoptosis and inhibit glucose metabolism by targeting PFKFB3 [78]. In addition, it has been illustrated that circSAMD4A and circ0014130 can significantly promote 5-Fu resistance in CRC cells by acting as miRNA sponges to upregulate PFKFB3 expression and lactate generation [79, 80]. The study by Huang et al. has shown evidence that transport circ0005963 deriving from oxaliplatin-resistant cells to susceptible cells can promote oxaliplatin resistance by sponging miR-122 to activate PKM2 [157]. Apart from circ0005963, lncRNA XIST is also implicated in sponge miR-137 to enhance the resistance of 5-Fu/cisplatin in CRC cells by maintaining a higher PKM2/PKM1 ratio [158]. HK2 is an essential enzyme for glycolysis, which has been elaborated to be targeted by the LncRNA DANCR/miR-125b-5p axis involved in regulating the glucose metabolism of CRC cells and inducing cisplatin resistance [159].

Fig. 4
figure 4

The mechanism of ncRNAs mediate chemotherapy resistance through metabolic reprogramming

A similar effect of ncRNAs on other metabolism reprogramming has been illustrated in vast research. It is demonstrated that increased expression of lncRNA MACC1-AS1 is documented to promote 5-Fu and oxaliplatin resistance by sponging miR-145-5p to inhibit the fatty acid oxidation pathway [160]. Furthermore, increased expression of miR-1291 is shown to target GLUT1-mediated glycolysis and ASS1-mediated arginine hydrolysis pathways, enhancing the susceptibility of prostate cancer (PC) cells to arginine deprivation and cisplatin therapy [161]. Li et al. have identified that re-expression of miR-34a in oxaliplatin-resistant CRC cells can increase their mRNA stability and restore sensitivity to oxaliplatin by targeting ornithine decarboxylase antizyme 2 (OAZ2) [162]. NcRNA involved in regulating metabolic reprogramming and chemoresistance will serve as novel anticancer strategies for cancer in the future. Nevertheless, the potential regulatory mechanism of ncRNAs mediating chemoresistance and metabolism balance needs to be further investigated.

NcRNAs mediate radiotherapy resistance through metabolic reprogramming

Dysregulate ncRNAs facilitate radiotherapy resistance by affecting metabolic reprogramming has been summarized in Fig. 5. Liang et al. have identified that lncRNA GLTC can promote radioiodine (RAI) resistance in papillary thyroid cancer by promoting LDHA enzymatic activity [163]. Increasing research has found that the radiotherapy resistance mediated by ncRNAs is associated with the AKT pathway. For instance, increased expression of lncRNA HCP5 has been identified by Guo et al. to induce radiotherapy resistance in esophageal cancer tissues by promoting AKT activation through regulating miR-216a-3p/PDK1 axis [164]. Another well-documented lncRNA DUXAP8 increases radiotherapy resistance in breast cancer cells by activating the PI3K/AKT/mTOR signaling pathway [165]. Moreover, the expression of SNHG7 is demonstrated to be upregulated in TC tissues and correlated with [131]I resistance as the increased expression of SNHG7 promotes [131]I resistance in TC cells by sponging miR-9-5p and increasing DPP4 expression to activate PI3K/AKT [166]. Taken together, dysregulated expression of ncRNAs induces the development of radiation therapy resistance in cancer cells mainly by targeting the PI3K/AKT signaling pathway. All the above findings have provided promising ncRNAs-targeted strategies for radiotherapy resistance.

Fig. 5
figure 5

The mechanism of ncRNAs mediate radiotherapy resistance through metabolic reprogramming

NcRNAs mediate immune resistance through metabolic reprogramming

Dysregulation of NcRNAs in metabolic networks is closely associated with immunotherapy tolerance and it has been summarized in Fig. 6. A currently published study has shown that overexpression of LINC01085 combined with immune checkpoint blockade is an effective strategy for the treatment of HIPC patients by inhibiting the interaction between TANK-binding kinase 1 (TBK1) and glycogen synthase kinase 3β (GSK3β) [167]. It is identified by Song et.al that lncRNA CASC11drives HCC progression by recruiting EIF4A3 to enhance E2F1 stability, which activates NF-κB signaling and PI3K/AKT/mTOR pathway and further upregulates glycolysis and the expression of PD-L1 [168]. As an emerging therapeutic approach, immunotherapy has been broadly applied in clinical therapy. Current findings suggest that the various combinations of treatments with produced anti-PD-1, CD80-Fc, and 4-1BBL-Fc proteins reduce inhibitory signals while increasing activatory signals, potentially lowering the risk of tumor resistance and improving treatment efficacy [169]. Based on previous studies, inhibiting NEAT1, XIST, and IL-6 may be very effective for the clinical treatment of GC [156, 170]. Recently, Elina Kaviani et.al revealed polyphenol natural compounds and maggot larvae (as sources for safe and effective bioactive compounds) can contribute to immunotherapy and chemotherapy activity as a complementary therapy via reducing the expression level of MKI67, CXCL8, IL-6/IL-6R, lncRNA XIST and NEAT1 and ultimately decreased the size and number of tumors [156]. Hence, the combination of ncRNAs and immunotherapy drugs is likely to be an effective strategy for improving immune treatment efficacy [167, 168, 171].

Fig. 6
figure 6

The mechanism of ncRNAs mediate immune and targeted resistance through metabolic reprogramming

NcRNAs mediate targeted therapy resistance through metabolic reprogramming

Dysregulation of ncRNAs mediates targeted therapy resistance through metabolic pathways has been summarized in Fig. 6. Han et al. have found that LncRNA AGAP2-AS1 can enhance breast cancer cell resistance to trastuzumab through miR-15a-5p/CPT1 axis to activate fatty acid oxidation (FAO) [172]. Liang et al. found that high expression of circ CDYL2 in breast cancer tissues conferred resistance to trastuzumab. Mechanically, circCDYL2 stabilizes GRB7 by preventing its ubiquitinated degradation and enhances its interaction with FAK, thereby maintaining downstream AKT and ERK1/2 activity to support trastuzumab resistance in breast cancer cells. However, HER2+ BC cells with high expression of circCDYL2 can reverse their resistance to trastuzumab by using FAK or GRB7 inhibitors [173]. The development of targeted therapeutic resistance is caused by dysregulated ncRNAs. To understand how metabolic reprogramming brought on by ncRNAs impacts cancer-targeted treatment, more study is required.

In conclusion, numerous studies have illustrated that dysregulated ncRNAs expression in cancer mediates the development of cancer therapeutic tolerance by regulating metabolic networks, and targeting dysregulated ncRNAs in cancer cells has great promise for overcoming cancer therapeutic tolerance, especially tolerance to chemotherapy treatment. More research is necessary to fully elucidate how ncRNAs influence cancer tolerance through metabolic reprogramming.

Targeting ncRNAs-mediated metabolic pathways provides effective strategies for cancer therapy

There are numerous studies have demonstrated that ncRNAs are involved in multiple metabolic pathways or signaling pathways to regulate the genesis and progression of cancer cells [174,175,176]. Therefore, ncRNAs-based metabolic reprogramming may provide promising prospects in overcoming cancer therapeutic tolerance and improving related anti-cancer drugs’ therapeutic efficiency. Elucidating the interaction and possible mechanism between ncRNAs and metabolic reprogramming has shed some insights into understanding the pathogenesis and provided a theoretical foundation for cancer research.

The treatments suppressing cancer progression through the metabolic networks mediated ncRNAs have been summarized in Fig. 7. Ni et al. revealed that simvastatin could decrease lncRNA SNHG29 expression and, thereby inhibit activation-induced PD-L1 expression, which enhances anti-cancer immunity [124]. Therefore, the use of cholesterol scavengers may significantly enhance the efficacy of immunotherapy [5]. Besides, numerous drugs affect ncRNA-mediated glucose reprogramming to suppress cancer progression. Wu et al. revealed that kaempferol reduced PKM2 expression and thereby inhibited cancer progression by upregulating miR-339-5p [177]. LncRNA UCA1 is documented to contribute to paclitaxel resistance and promote glycolysis by facilitating the expression of HK2 and LDHA [178]. LncRNA FGD5-AS1 promotes glycolysis and 5-FU resistance of CRC cells by acting as a ceRNA for miR-330-3p and upregulating HK2 in CRC. While erlotinib could effectively re-sensitivity CRC to 5-Fu by targeting lncRNA FGD5-AS1 [179].

Fig. 7
figure 7

The mechanism of treatments affecting cancer progression by targeting the dysregulated ncRNAs mediated metabolic networks

Importantly, targeting dysregulated ncRNAs by delivering small molecule drugs to the body provides a novel approach for future cancer therapy. Antisense oligonucleotides (ASO) of lncRNA AGPG delivered to patients with ESCC have been demonstrated to effectively limit glycolysis and cell proliferation to inhibit the expression of PFKFB3 [180]. Song et al. demonstrated that siRNA specific to lncRNA HISLA inhibits glycolysis and chemoresistance in breast cancer by mediating the interaction of HIF-1α with prolyl hydroxylase 2 (PHD2) [181]. The study by Yun et al. has identified that applicate inhibition of LINRIS via in vivo-optimized RNA interference (RNAi) and oxaliplatin could effectively suppress glycolysis and cell proliferation in CRC [46]. The reprogramming of energy metabolism mediated by ncRNAs is a potential target for some drugs [124, 177,178,179]. As a novel therapeutic approach, the combination of small molecule inhibitors with cancer therapy can significantly suppress the progression of cancer [180, 181]. However, the main challenge of their application currently lies in how to precisely target specific cell types. More notably, despite the rapid development of CAR-T cell therapy in non-solid cancers, its treatment currently faces many challenges [182]. Whether lncRNAs can serve as ideal targets to provide new strategies to improve the efficiency of CAR-T cell immune treatment deserves further investigation.

Conclusion

Energy metabolic reprogramming is a fundamental feature of cancers. Cancer cells reprogram substances such as glucose, lipid and amino acids to satisfy their needs for rapid proliferation and adaptation to external stress, which is driven by dysregulated ncRNAs. Interestingly, numerous studies have suggested that ncRNAs derived from gut microbial and viruses are involved in metabolic reprogramming which may be an emerging direction for future research. Importantly, sufficient studies have demonstrated that alterations in metabolic pathways mediated by ncRNAs are closely associated with the development of cancer treatment tolerance, therefore, the targeted ncRNAs therapy in combination with conventional cancer therapy may be an effective strategy to overcome low response or tolerance of cancer therapy. However, current studies on the metabolism of ncRNAs are mainly focused on glucose and lipids, whereas molecular mechanisms in amino acid metabolism and more systematic research are still required. Moreover, the issues about how to target ncRNAs in specific cells to improve the efficacy of treatment need to be further researched. In conclusion, this study emphasizes the feasibility of targeting lncRNAs and presents new insights into the molecular signaling pathways associated with metabolism.

Availability of data and materials

Not applicable.

Abbreviations

3'-UTR:

3’-Untranslated regions

5-Fu:

5-Fluorouracil

ACAC:

Acetyl coenzyme A carboxylase

ACAD9:

Acyl coenzyme a dehydrogenase-9

ACC:

Acetyl coenzyme A carboxylase

ACLY:

ATP-citrate synthase

AMPK:

AMP-activated protein kinase

ASAP:

ATP synthase-associated peptide

ASCL2:

Long-chain-fatty-acid-CoA ligase 2

ASCT2:

Amino-acid transporter 2

ASO:

Antisense oligonucleotides

ASS1:

Argininosuccinate synthase 1

ATP:

Adenosine triphosphate

BCL6:

B-cell lymphoma 6 protein

cccDNA:

Covalently closed loop DNA

ceRNA:

Competitive endogenous RNA

CircRNAs:

Circular non-coding RNAs

CPT-1:

Carnitine palmityltransferase-1

CRC:

Colorectal cancer

ENO1:

Alpha-enolase

EVs:

Vesicles

FABPs:

Fatty acid binding proteins

FAO:

Fatty acid oxidation

FASN:

Fatty acid synthase

FATPs:

Fatty acid transport proteins

G6PD:

Glucose-6-phosphate dehydrogenase

GC:

Gastric cancer

GLS:

Glutaminase

GLUT1:

Glucose transporter1

GPT2:

Glutamic–pyruvic transaminase

GSH:

Glutathione

GSK3β:

Glycogen synthase kinase 3β

HBV:

Hepatitis B virus

HCC:

Hepatocellular carcinoma

HIF-1α:

Hypoxia-inducible factor-1α

HK2:

Hexokinase2

HMGCR:

HMG-CoA reductase

HSF1:

Heat shock factor 1

HSP:

Heat shock protein

IDO1:

Indoleamine 2, 3-dioxygenase 1

IFN-γ:

Interferon-γ

LDHA:

Lactate dehydrogenase A

LDLRs:

Low-density lipoproteinreceptor superfamily

LncRNAs:

Long non-coding RNAs

M1:

Classical activated macrophages

M2:

Alternatively activated macrophages

MDSCs:

Myeloid-derived suppressor cells

miRNA:

MicroRNA

mRNA:

Messenger RNA

MSCs:

Mesenchymal stem cells

mtDNA:

Mitochondrial DNA

mTORC:

Mammalian target of rapamycin

NAFLD:

Non-alcoholic fatty liver disease

NcRNAs:

Non-coding RNAs

NK:

Natural killer

NPC:

Nasopharyngeal carcinoma

NRF:

Nuclear respiratory factor

OAZ2:

Ornithine decarboxylase antizyme 2

ODC1:

Ornithine decarboxylase 1

OXPHOS:

Oxidative phosphorylation

PC:

Prostate cancer

PDC:

Pyruvate dehydrogenase complex

PDK:

Pyruvate dehydrogenase kinase

PD-L1:

Programmed death ligand-1

PFK-1:

Phosphofructokinase-1

PFKFB3:

6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3

PGC-1α:

PPARγ coactivator-1α

PGK1:

Phosphoglycerate kinase 1

PHD2:

Prolyl hydroxylase 2

PHGDH:

Phosphoglycerate dehydrogenase

piRNA:

PIWI-interacting RNA

PKM2:

Pyruvate kinase M2

PPAR:

Peroxisome proliferators-activated receptors

PPP:

Pentose phosphate pathway

PTEN:

Phosphatase and tensin homolog

PTMs:

Post-transcriptional modifications

RAI:

Radioiodine

RNAi:

RNA interference

ROS:

Reactive oxygen species

rRNA:

Ribosomal RNA

SCD:

Stearoyl-CoA desaturase

SDH:

Succinate dehydrogenase

snoRNA:

Small nucleolar RNA

snRNA:

Small nuclear RNA

SQLE:

Squalene monooxygenase

SREBP:

Sterol-regulatory element binding proteins

SRSF3:

Serine and arginine-rich splicing factor 3

STAT3:

Signal transducer and activator of transcription 3

TAC:

Tricarboxylic acid cycle

TBK1:

TANK-binding kinase 1

TDO:

Tryptophan 2,3-dioxygenase

TFAM:

Mitochondrial transcription factor A

TME:

Tumor microenvironment

TNBC:

Triple-negative breast cancer

tRNA:

Transfer RNA

VLDLR:

Very low-density-lipoprotein receptor

YAP:

Yes-Associated Protein

References

  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73:17–48. https://doi.org/10.3322/caac.21763.

    Article  PubMed  Google Scholar 

  2. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31–46. https://doi.org/10.1158/2159-8290.Cd-21-1059.

    Article  CAS  PubMed  Google Scholar 

  3. van Weverwijk A, de Visser KE. Mechanisms driving the immunoregulatory function of cancer cells. Nat Rev Cancer. 2023. https://doi.org/10.1038/s41568-022-00544-4.

    Article  PubMed  Google Scholar 

  4. Zhang X, Ji L, Li MO. Control of tumor-associated macrophage responses by nutrient acquisition and metabolism. Immunity. 2023;56:14–31. https://doi.org/10.1016/j.immuni.2022.12.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang Q, et al. Regulation of PD-L1 through direct binding of cholesterol to CRAC motifs. Sci Adv. 2022;8:4722. https://doi.org/10.1126/sciadv.abq4722.

    Article  CAS  Google Scholar 

  6. Byun JK, et al. Inhibition of glutamine utilization synergizes with immune checkpoint inhibitor to promote antitumor immunity. Mol Cell. 2020;80(592–606):e598. https://doi.org/10.1016/j.molcel.2020.10.015.

    Article  CAS  Google Scholar 

  7. Yan S, et al. Emerging role of non-coding RNAs in glucose metabolic reprogramming and chemoresistance in colorectal cancer. Front Oncol. 2022;12:954329. https://doi.org/10.3389/fonc.2022.954329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fathi M, Ghafouri-Fard S, Abak A, Taheri M. Emerging roles of miRNAs in the development of pancreatic cancer. Biomed Pharmacother. 2021;141:111914. https://doi.org/10.1016/j.biopha.2021.111914.

    Article  CAS  PubMed  Google Scholar 

  9. Cai ZR, et al. Circular RNAs: emerging regulators of glucose metabolism in cancer. Cancer Lett. 2023;552:215978. https://doi.org/10.1016/j.canlet.2022.215978.

    Article  CAS  PubMed  Google Scholar 

  10. Nagao A, Kobayashi M, Koyasu S, Chow CCT, Harada H. HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int J Mol Sci. 2019. https://doi.org/10.3390/ijms20020238.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Camarda R, Williams J, Goga A. In vivo reprogramming of cancer metabolism by MYC. Front Cell Dev Biol. 2017;5:35. https://doi.org/10.3389/fcell.2017.00035.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Liu J, Zhang C, Hu W, Feng Z. Tumor suppressor p53 and metabolism. J Mol Cell Biol. 2019;11:284–92. https://doi.org/10.1093/jmcb/mjy070.

    Article  CAS  PubMed  Google Scholar 

  13. Zhang X, et al. Transcriptional repression of aerobic glycolysis by OVOL2 in breast cancer. Adv Sci. 2022;9:e2200705. https://doi.org/10.1002/advs.202200705.

    Article  CAS  Google Scholar 

  14. Nagel AK, Ball LE. Intracellular protein O-GlcNAc modification integrates nutrient status with transcriptional and metabolic regulation. Adv Cancer Res. 2015;126:137–66. https://doi.org/10.1016/bs.acr.2014.12.003.

    Article  CAS  PubMed  Google Scholar 

  15. Yang Y, et al. O-GlcNAcylation of YTHDF2 promotes HBV-related hepatocellular carcinoma progression in an N(6)-methyladenosine-dependent manner. Signal Transduct Target Ther. 2023;8:63. https://doi.org/10.1038/s41392-023-01316-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yuan Y, et al. Cancer metabolism and tumor microenvironment: fostering each other? Sci China Life Sci. 2022;65:236–79. https://doi.org/10.1007/s11427-021-1999-2.

    Article  CAS  PubMed  Google Scholar 

  17. Corbet C, Feron O. Tumour acidosis: from the passenger to the driver’s seat. Nat Rev Cancer. 2017;17:577–93. https://doi.org/10.1038/nrc.2017.77.

    Article  CAS  PubMed  Google Scholar 

  18. Gu J, et al. Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep. 2022;39:110986. https://doi.org/10.1016/j.celrep.2022.110986.

    Article  CAS  PubMed  Google Scholar 

  19. Bian X, et al. Lipid metabolism and cancer. J Exp Med. 2021. https://doi.org/10.1084/jem.20201606.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun. 2018. https://doi.org/10.1186/s40880-018-0301-4.

    Article  Google Scholar 

  21. Du D, et al. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B. 2022;12:558–80. https://doi.org/10.1016/j.apsb.2021.09.019.

    Article  CAS  PubMed  Google Scholar 

  22. Zhang Z, et al. DHHC9-mediated GLUT1 S-palmitoylation promotes glioblastoma glycolysis and tumorigenesis. Nat Commun. 2021;12:5872. https://doi.org/10.1038/s41467-021-26180-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Qin Y, Qiao Y, Wang D, Tang C, Yan G. Ferritinophagy and ferroptosis in cardiovascular disease: mechanisms and potential applications. Biomed Pharmacother. 2021;141:111872. https://doi.org/10.1016/j.biopha.2021.111872.

    Article  CAS  PubMed  Google Scholar 

  24. Xiang H, et al. Metabolic reprogramming of immune cells in pancreatic cancer progression. Biomed Pharmacother. 2023;157:113992. https://doi.org/10.1016/j.biopha.2022.113992.

    Article  CAS  PubMed  Google Scholar 

  25. Wu L, et al. RIPK3 orchestrates fatty acid metabolism in tumor-associated macrophages and hepatocarcinogenesis. Cancer Immunol Res. 2020;8:710–21. https://doi.org/10.1158/2326-6066.CIR-19-0261.

    Article  CAS  PubMed  Google Scholar 

  26. Goossens P, et al. Membrane cholesterol efflux drives tumor-associated macrophage reprogramming and tumor progression. Cell Metab. 2019;29:1376-1389.e1374. https://doi.org/10.1016/j.cmet.2019.02.016.

    Article  CAS  PubMed  Google Scholar 

  27. Wang D, Wan X. Progress in research on the role of amino acid metabolic reprogramming in tumour therapy: a review. Biomed Pharmacother. 2022;156:113923. https://doi.org/10.1016/j.biopha.2022.113923.

    Article  CAS  PubMed  Google Scholar 

  28. Li T, Le A. Glutamine metabolism in cancer. Adv Exp Med Biol. 2018;1063:13–32. https://doi.org/10.1007/978-3-319-77736-8_2.

    Article  CAS  PubMed  Google Scholar 

  29. Shan X, et al. Serine metabolism orchestrates macrophage polarization by regulating the IGF1-p38 axis. Cell Mol Immunol. 2022;19:1263–78. https://doi.org/10.1038/s41423-022-00925-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Newman AC, et al. Immune-regulated IDO1-dependent tryptophan metabolism is source of one-carbon units for pancreatic cancer and stellate cells. Mol Cell. 2021;81:2290–302. https://doi.org/10.1016/j.molcel.2021.03.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cronin SJF, et al. The metabolite BH4 controls T cell proliferation in autoimmunity and cancer. Nature. 2018;563:564–8. https://doi.org/10.1038/s41586-018-0701-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang W, et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569:270–4. https://doi.org/10.1038/s41586-019-1170-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. Sci Adv. 2016;2:e1600200. https://doi.org/10.1126/sciadv.1600200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Elia I, Haigis MC. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab. 2021;3:21–32. https://doi.org/10.1038/s42255-020-00317-z.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018;18:5–18. https://doi.org/10.1038/nrc.2017.99.

    Article  CAS  PubMed  Google Scholar 

  36. Zhang Q, et al. miR-4999-5p predicts colorectal cancer survival outcome and reprograms glucose metabolism by targeting PRKAA2. Onco Targets Ther. 2020;13:1199–210. https://doi.org/10.2147/ott.S234666.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yang W, et al. Exosomal miR-205-5p enhances angiogenesis and nasopharyngeal carcinoma metastasis by targeting desmocollin-2. Mol Ther Oncolytics. 2022;24:612–23. https://doi.org/10.1016/j.omto.2022.02.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yu B, et al. Post-transcriptional regulation of tumor suppressor gene lncRNA CARMN via m(6)A modification and miRNA regulation in cervical cancer. J Cancer Res Clin Oncol. 2023. https://doi.org/10.1007/s00432-023-04893-x.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Huang CJ, Choo KB. Circular RNA- and microRNA-mediated post-transcriptional regulation of preadipocyte differentiation in adipogenesis: from expression profiling to signaling pathway. Int J Mol Sci. 2023. https://doi.org/10.3390/ijms24054549.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Li Y, et al. Non-coding RNA in bladder cancer. Cancer Lett. 2020;485:38–44. https://doi.org/10.1016/j.canlet.2020.04.023.

    Article  CAS  PubMed  Google Scholar 

  41. Liao W, et al. The role and mechanism of noncoding RNAs in regulation of metabolic reprogramming in hepatocellular carcinoma. Int J Cancer. 2022;151:337–47. https://doi.org/10.1002/ijc.34040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509–24. https://doi.org/10.1038/nrm3838.

    Article  CAS  PubMed  Google Scholar 

  43. Ali Syeda Z, Langden SSS, Munkhzul C, Lee M, Song SJ. Regulatory mechanism of MicroRNA expression in cancer. Int J Mol Sci. 2020. https://doi.org/10.3390/ijms21051723.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Cabili MN, et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011;25:1915–27. https://doi.org/10.1101/gad.17446611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22:96–118. https://doi.org/10.1038/s41580-020-00315-9.

    Article  CAS  PubMed  Google Scholar 

  46. Wang Y, et al. LncRNA LINRIS stabilizes IGF2BP2 and promotes the aerobic glycolysis in colorectal cancer. Mol Cancer. 2019;18:174. https://doi.org/10.1186/s12943-019-1105-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Liu X, et al. LINC00839 promotes colorectal cancer progression by recruiting RUVBL1/Tip60 complexes to activate NRF1. EMBO Rep. 2022;23:e54128. https://doi.org/10.15252/embr.202154128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen LL. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17:205–11. https://doi.org/10.1038/nrm.2015.32.

    Article  CAS  PubMed  Google Scholar 

  49. Zhou WY, et al. Circular RNA: metabolism, functions and interactions with proteins. Mol Cancer. 2020;19:172. https://doi.org/10.1186/s12943-020-01286-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ma J, et al. CircEGFR reduces the sensitivity of pirarubicin and regulates the malignant progression of triple-negative breast cancer via the miR-1299/EGFR axis. Int J Biol Macromol. 2023. https://doi.org/10.1016/j.ijbiomac.2023.125295.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Fang X, et al. LncRNA SNHG1 knockdown inhibits hyperglycemia induced ferroptosis via miR-16-5p/ACSL4 axis to alleviate diabetic nephropathy. J Diabetes Invest. 2023. https://doi.org/10.1111/jdi.14036.

    Article  Google Scholar 

  52. Safi A, et al. The role of noncoding RNAs in metabolic reprogramming of cancer cells. Cell Mol Biol Lett. 2023;28:37. https://doi.org/10.1186/s11658-023-00447-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tan S, et al. Exosomal cargos-mediated metabolic reprogramming in tumor microenvironment. J Exp Clin Cancer Res. 2023;42:59. https://doi.org/10.1186/s13046-023-02634-z.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Yang H, et al. miRNA-874-3p inhibits the migration, invasion and proliferation of breast cancer cells by targeting VDAC1. Aging. 2023;15:705–17. https://doi.org/10.18632/aging.204474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhou Y, et al. miR-125b-5p suppresses leukemia cell proliferation by regulating MCL1. J Environ Pathol Toxicol Oncol. 2023;42:17–26. https://doi.org/10.1615/JEnvironPatholToxicolOncol.2022041924.

    Article  PubMed  Google Scholar 

  56. Li X, et al. MicroRNA-1205 suppresses hepatocellular carcinoma cell proliferation via a CSNK2B/CDK4 axis. Technol Cancer Res Treat. 2023. https://doi.org/10.1177/15330338221150544.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Chen C, Zhang Q, Kong B. miRNA-576-5p promotes endometrial cancer cell growth and metastasis by targeting ZBTB4. Clin Transl Oncol. 2023;25:706–20. https://doi.org/10.1007/s12094-022-02976-8.

    Article  CAS  PubMed  Google Scholar 

  58. Li L, Liu P, He C, Xu C. miRNA-223-3p regulates ECT2 to promote proliferation, invasion, and metastasis of gastric cancer through the Wnt/β-catenin signaling pathway. J Cancer Res Clin Oncol. 2023;149:121–34. https://doi.org/10.1007/s00432-022-04453-9.

    Article  CAS  PubMed  Google Scholar 

  59. He J, et al. Circular RNA circHERC4 as a novel oncogenic driver to promote tumor metastasis via the miR-556-5p/CTBP2/E-cadherin axis in colorectal cancer. J Hematol Oncol. 2021;14:194. https://doi.org/10.1186/s13045-021-01210-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Liu S, et al. SP1-induced lncRNA DUBR promotes stemness and oxaliplatin resistance of hepatocellular carcinoma via E2F1-CIP2A feedback. Cancer Lett. 2022;528:16–30. https://doi.org/10.1016/j.canlet.2021.12.026.

    Article  CAS  PubMed  Google Scholar 

  61. Chen J, et al. Tumor-derived exosomal miR-19b-3p facilitates M2 macrophage polarization and exosomal LINC00273 secretion to promote lung adenocarcinoma metastasis via Hippo pathway. Clin Transl Med. 2021;11:e478. https://doi.org/10.1002/ctm2.478.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Chong X, et al. PIK3CA mutations-mediated downregulation of circLHFPL2 inhibits colorectal cancer progression via upregulating PTEN. Mol Cancer. 2022;21:118. https://doi.org/10.1186/s12943-022-01531-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Deng F, et al. Tumor-secreted dickkopf2 accelerates aerobic glycolysis and promotes angiogenesis in colorectal cancer. Theranostics. 2019;9:1001–14. https://doi.org/10.7150/thno.30056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Song L, Dong N, Li Z. p, p’-Dichlorodiphenyltrichloroethane promotes aerobic glycolysis via reactive oxygen species-mediated extracellular signal-regulated kinase/M2 isoform of pyruvate kinase (PKM2) signaling in colorectal cancer cells. Environ Toxicol. 2020;35:333–45. https://doi.org/10.1002/tox.22869.

    Article  CAS  PubMed  Google Scholar 

  65. Wang H, et al. The LncRNA FEZF1-AS1 promotes tumor proliferation in colon cancer by regulating the mitochondrial protein PCK2. Oncol Res. 2021;29:201–15. https://doi.org/10.32604/or.2022.03553.

    Article  PubMed  Google Scholar 

  66. Bergamini C, et al. MiR-494 induces metabolic changes through G6pc targeting and modulates sorafenib response in hepatocellular carcinoma. J Exp Clin Cancer Res. 2023;42:145. https://doi.org/10.1186/s13046-023-02718-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wei Z, Cui L, Mei Z, Liu M, Zhang D. miR-181a mediates metabolic shift in colon cancer cells via the PTEN/AKT pathway. FEBS Lett. 2014;588:1773–9. https://doi.org/10.1016/j.febslet.2014.03.037.

    Article  CAS  PubMed  Google Scholar 

  68. Shi Y, et al. Let-7a-5p inhibits triple-negative breast tumor growth and metastasis through GLUT12-mediated warburg effect. Cancer Lett. 2020;495:53–65. https://doi.org/10.1016/j.canlet.2020.09.012.

    Article  CAS  PubMed  Google Scholar 

  69. Mo Y, et al. Circular RNA circRNF13 inhibits proliferation and metastasis of nasopharyngeal carcinoma via SUMO2. Mol Cancer. 2021;20:112. https://doi.org/10.1186/s12943-021-01409-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li B, et al. LncRNA GAL promotes colorectal cancer liver metastasis through stabilizing GLUT1. Oncogene. 2022;41:1882–94. https://doi.org/10.1038/s41388-022-02230-z.

    Article  CAS  PubMed  Google Scholar 

  71. Zhang ZJ, Zhang YH, Qin XJ, Wang YX, Fu J. Circular RNA circDENND4C facilitates proliferation, migration and glycolysis of colorectal cancer cells through miR-760/GLUT1 axis. Eur Rev Med Pharmacol Sci. 2020;24:2387–400. https://doi.org/10.26355/eurrev_202003_20506.

    Article  PubMed  Google Scholar 

  72. Cui Y, et al. m(6)A-modified circFOXK2 targets GLUT1 to accelerate oral squamous cell carcinoma aerobic glycolysis. Cancer Gene Ther. 2023;30:163–71. https://doi.org/10.1038/s41417-022-00526-6.

    Article  CAS  PubMed  Google Scholar 

  73. Chen C, et al. Long noncoding RNA KCNQ1OT1 promotes colorectal carcinogenesis by enhancing aerobic glycolysis via hexokinase-2. Aging. 2020;12:11685–97. https://doi.org/10.18632/aging.103334.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhao S, et al. LncRNA MIR17HG promotes colorectal cancer liver metastasis by mediating a glycolysis-associated positive feedback circuit. Oncogene. 2021;40:4709–24. https://doi.org/10.1038/s41388-021-01859-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Xiong X, et al. High-risk pathological subtype associated FAM83A-AS1 promotes malignancy and glycolysis of lung adenocarcinoma via miR-202-3p/HK2 axis. Oncol Rep. 2023. https://doi.org/10.3892/or.2023.8532.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Liu Y, et al. Hsa_circ_0000231 knockdown inhibits the glycolysis and progression of colorectal cancer cells by regulating miR-502-5p/MYO6 axis. World J Surg Oncol. 2020;18:255. https://doi.org/10.1186/s12957-020-02033-0.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Cui S, Yang X, Zhang L, Zhao Y, Yan W. LncRNA MAFG-AS1 promotes the progression of colorectal cancer by sponging miR-147b and activation of NDUFA4. Biochem Biophys Res Commun. 2018;506:251–8. https://doi.org/10.1016/j.bbrc.2018.10.112.

    Article  CAS  PubMed  Google Scholar 

  78. Deng X, et al. Mir-488 alleviates chemoresistance and glycolysis of colorectal cancer by targeting PFKFB3. J Clin Lab Anal. 2021;35:e23578. https://doi.org/10.1002/jcla.23578.

    Article  CAS  PubMed  Google Scholar 

  79. Gao Y, Liu C, Xu X, Wang Y, Jiang Y. Circular RNA sterile alpha motif domain containing 4A contributes to cell 5-fluorouracil resistance in colorectal cancer by regulating the miR-545-3p/6-phosphofructo-2-kinase/fructose-2,6-bisphosphataseisotype 3 axis. Anticancer Drugs. 2022;33:553–63. https://doi.org/10.1097/cad.0000000000001285.

    Article  CAS  PubMed  Google Scholar 

  80. Wang W, Zhou L, Li Z, Lin G. Circ_0014130 is involved in the drug sensitivity of colorectal cancer through miR-197-3p/PFKFB3 axis. J Gastroenterol Hepatol. 2022;37:908–18. https://doi.org/10.1111/jgh.15829.

    Article  CAS  PubMed  Google Scholar 

  81. Zhang S, et al. Fusobacterium nucleatum promotes epithelial-mesenchymal transiton through regulation of the lncRNA MIR4435-2HG/miR-296-5p/Akt2/SNAI1 signaling pathway. Febs j. 2020;287:4032–47. https://doi.org/10.1111/febs.15233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Hong XL, et al. Metformin abrogates Fusobacterium nucleatum-induced chemoresistance in colorectal cancer by inhibiting miR-361-5p/sonic hedgehog signaling-regulated stemness. Br J Cancer. 2023;128:363–74. https://doi.org/10.1038/s41416-022-02044-6.

    Article  CAS  PubMed  Google Scholar 

  83. Hong J, et al. F. nucleatum targets lncRNA ENO1-IT1 to promote glycolysis and oncogenesis in colorectal cancer. Gut. 2021;70:2123–37. https://doi.org/10.1136/gutjnl-2020-322780.

    Article  CAS  PubMed  Google Scholar 

  84. Ma J, et al. Long non-coding RNA ANRIL promotes chemoresistance in triple-negative breast cancer via enhancing aerobic glycolysis. Life Sci. 2022;306:120810. https://doi.org/10.1016/j.lfs.2022.120810.

    Article  CAS  PubMed  Google Scholar 

  85. Wang L, et al. Exosome-transferred LINC01559 promotes the progression of gastric cancer via PI3K/AKT signaling pathway. Cell Death Dis. 2020;11:723. https://doi.org/10.1038/s41419-020-02810-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Fu R, Yang P, Amin S, Li Z. A novel miR-206/hnRNPA1/PKM2 axis reshapes the Warburg effect to suppress colon cancer growth. Biochem Biophys Res Commun. 2020;531:465–71. https://doi.org/10.1016/j.bbrc.2020.08.019.

    Article  CAS  PubMed  Google Scholar 

  87. Lan Z, et al. 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. 2020;10:363. https://doi.org/10.3389/fonc.2020.00363.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Huang JZ, et al. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 2017;68:171–84. https://doi.org/10.1016/j.molcel.2017.09.015.

    Article  CAS  PubMed  Google Scholar 

  89. Bian Z, et al. LncRNA-FEZF1-AS1 promotes tumor proliferation and metastasis in colorectal cancer by regulating PKM2 signaling. Clin Cancer Res. 2018;24:4808–19. https://doi.org/10.1158/1078-0432.Ccr-17-2967.

    Article  CAS  PubMed  Google Scholar 

  90. Hua Q, et al. Hypoxia-induced lncRNA-AC020978 promotes proliferation and glycolytic metabolism of non-small cell lung cancer by regulating PKM2/HIF-1alpha axis. Theranostics. 2020;10:4762–78. https://doi.org/10.7150/thno.43839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhang Z, et al. LncRNA MCF2L-AS1 aggravates proliferation, invasion and glycolysis of colorectal cancer cells via the crosstalk with miR-874-3p/FOXM1 signaling axis. Carcinogenesis. 2021;42:263–71. https://doi.org/10.1093/carcin/bgaa093.

    Article  CAS  PubMed  Google Scholar 

  92. Tang J, et al. LncRNA GLCC1 promotes colorectal carcinogenesis and glucose metabolism by stabilizing c-Myc. Nat Commun. 2019;10:3499. https://doi.org/10.1038/s41467-019-11447-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhu Y, et al. The long noncoding RNA glycoLINC assembles a lower glycolytic metabolon to promote glycolysis. Mol Cell. 2022;82:542–54. https://doi.org/10.1016/j.molcel.2021.11.017.

    Article  CAS  PubMed  Google Scholar 

  94. Zhang Y, et al. LINC01615 maintains cell survival in adaptation to nutrient starvation through the pentose phosphate pathway and modulates chemosensitivity in colorectal cancer. Cell Mol Life Sci. 2022;80:20. https://doi.org/10.1007/s00018-022-04675-7.

    Article  CAS  PubMed  Google Scholar 

  95. Chen B, et al. N6-methyladenosine modification of circ_0003215 suppresses the pentose phosphate pathway and malignancy of colorectal cancer through the miR-663b/DLG4/G6PD axis. Cell Death Dis. 2022;13:804. https://doi.org/10.1038/s41419-022-05245-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wang X, Chen K, Zhao Z. LncRNA OR3A4 regulated the growth of osteosarcoma cells by modulating the miR-1207-5p/G6PD signaling. Onco Targets Ther. 2020;13:3117–28. https://doi.org/10.2147/ott.S234514.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Wang X, et al. lncRNA-encoded pep-AP attenuates the pentose phosphate pathway and sensitizes colorectal cancer cells to Oxaliplatin. EMBO Rep. 2022;23:e53140. https://doi.org/10.15252/embr.202153140.

    Article  CAS  PubMed  Google Scholar 

  98. Yang X, et al. LncRNA PDIA3P interacts with c-Myc to regulate cell proliferation via induction of pentose phosphate pathway in multiple myeloma. Biochem Biophys Res Commun. 2018;498:207–13. https://doi.org/10.1016/j.bbrc.2018.02.211.

    Article  CAS  PubMed  Google Scholar 

  99. Anwar S, Shamsi A, Mohammad T, Islam A, Hassan MI. Targeting pyruvate dehydrogenase kinase signaling in the development of effective cancer therapy. Biochim Et Biophys Acta Rev Cancer. 2021;1876:188568. https://doi.org/10.1016/j.bbcan.2021.188568.

    Article  CAS  Google Scholar 

  100. Cui Z, Wang Q, Deng MH, Han QL. LncRNA HCG11 promotes 5-FU resistance of colon cancer cells through reprogramming glucose metabolism by targeting the miR-144-3p-PDK4 axis. Cancer Biomark. 2022;34:41–53. https://doi.org/10.3233/cbm-210212.

    Article  CAS  PubMed  Google Scholar 

  101. Liu S, et al. SPRY4-IT1 promotes survival of colorectal cancer cells through regulating PDK1-mediated glycolysis. Anim Cells Syst. 2020;24:220–7. https://doi.org/10.1080/19768354.2020.1784274.

    Article  CAS  Google Scholar 

  102. Liu S, et al. miR-142-5p promotes development of colorectal cancer through targeting SDHB and facilitating generation of aerobic glycolysis. Biomed Pharmacother. 2017;92:1119–27. https://doi.org/10.1016/j.biopha.2017.05.134.

    Article  CAS  PubMed  Google Scholar 

  103. Liu YD, et al. Let-7a regulates EV secretion and mitochondrial oxidative phosphorylation by targeting SNAP23 in colorectal cancer. J Exp Clin Cancer Res. 2021;40:31. https://doi.org/10.1186/s13046-020-01813-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Collu-Marchese M, Shuen M, Pauly M, Saleem A, Hood DA. The regulation of mitochondrial transcription factor A (Tfam) expression during skeletal muscle cell differentiation. 2015. Biosci Rep. https://doi.org/10.1042/BSR20150073.

  105. Ge Q, et al. Micropeptide ASAP encoded by LINC00467 promotes colorectal cancer progression by directly modulating ATP synthase activity. J Clin Invest. 2021. https://doi.org/10.1172/JCI152911.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Sun L, et al. RNA-binding protein RALY reprogrammes mitochondrial metabolism via mediating miRNA processing in colorectal cancer. Gut. 2021;70:1698–712. https://doi.org/10.1136/gutjnl-2020-320652.

    Article  CAS  PubMed  Google Scholar 

  107. Zhao Y, et al. Nuclear-encoded lncRNA MALAT1 epigenetically controls metabolic reprogramming in HCC cells through the mitophagy pathway. Mol Ther Nucleic Acids. 2021;23:264–76. https://doi.org/10.1016/j.omtn.2020.09.040.

    Article  CAS  PubMed  Google Scholar 

  108. Currie E, Schulze A, Zechner R, Walther TC, Farese RV Jr. Cellular fatty acid metabolism and cancer. Cell Metab. 2013;18:153–61. https://doi.org/10.1016/j.cmet.2013.05.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Röhrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016;16:732–49. https://doi.org/10.1038/nrc.2016.89.

    Article  CAS  PubMed  Google Scholar 

  110. Zhang X, Yao J, Shi H, Gao B, Zhang L. LncRNA TINCR/microRNA-107/CD36 regulates cell proliferation and apoptosis in colorectal cancer via PPAR signaling pathway based on bioinformatics analysis. Biol Chem. 2019;400:663–75. https://doi.org/10.1515/hsz-2018-0236.

    Article  CAS  PubMed  Google Scholar 

  111. Christensen LL, et al. SNHG16 is regulated by the Wnt pathway in colorectal cancer and affects genes involved in lipid metabolism. Mol Oncol. 2016;10:1266–82. https://doi.org/10.1016/j.molonc.2016.06.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Gharib E, Nasri Nasrabadi P, Reza Zali M. miR-497–5p mediates starvation-induced death in colon cancer cells by targeting acyl-CoA synthetase-5 and modulation of lipid metabolism. J Cell Physiol. 2020;235:5570–89. https://doi.org/10.1002/jcp.29488.

    Article  CAS  PubMed  Google Scholar 

  113. Kim BK, Yoo HI, Lee AR, Choi K, Yoon SK. Decreased expression of VLDLR is inversely correlated with miR-200c in human colorectal cancer. Mol Carcinog. 2017;56:1620–9. https://doi.org/10.1002/mc.22618.

    Article  CAS  PubMed  Google Scholar 

  114. Mosapour A, Karami Tehrani FS, Atri M. Expression level of VLDL receptor and VLDL-c levels in the malignant and benign breast tumors: the correlation with miRNA-4465 and miRNA-1297. Mol Cell Probes. 2020;53:101624. https://doi.org/10.1016/j.mcp.2020.101624.

    Article  CAS  PubMed  Google Scholar 

  115. Liu F, et al. Long non-coding RNA SNHG6 couples cholesterol sensing with mTORC1 activation in hepatocellular carcinoma. Nat Metab. 2022;4:1022–40. https://doi.org/10.1038/s42255-022-00616-7.

    Article  CAS  PubMed  Google Scholar 

  116. Sallam T, et al. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat Med. 2018;24:304–12. https://doi.org/10.1038/nm.4479.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hu YW, et al. A lincRNA-DYNLRB2-2/GPR119/GLP-1R/ABCA1-dependent signal transduction pathway is essential for the regulation of cholesterol homeostasis. J Lipid Res. 2014;55:681–97. https://doi.org/10.1194/jlr.M044669.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Yang Y, et al. circCAPRIN1 interacts with STAT2 to promote tumor progression and lipid synthesis via upregulating ACC1 expression in colorectal cancer. Cancer Commun. 2023;43:100–22. https://doi.org/10.1002/cac2.12380.

    Article  Google Scholar 

  119. Peng JY, et al. Upregulation of superenhancer-driven LncRNA FASRL by USF1 promotes de novo fatty acid biosynthesis to exacerbate hepatocellular carcinoma. Adv Sci. 2022. https://doi.org/10.1002/advs.202204711.

    Article  Google Scholar 

  120. Meng Q, et al. Epstein-barr virus-encoded MicroRNA-BART18–3p promotes colorectal cancer progression by targeting de novo lipogenesis. Adv Sci. 2022;9:e2202116. https://doi.org/10.1002/advs.202202116.

    Article  CAS  Google Scholar 

  121. Wang X, et al. MYC-regulated mevalonate metabolism maintains brain tumor-initiating cells. Can Res. 2017;77:4947–60. https://doi.org/10.1158/0008-5472.CAN-17-0114.

    Article  CAS  Google Scholar 

  122. He M, et al. Pro-inflammation NF-kappaB signaling triggers a positive feedback via enhancing cholesterol accumulation in liver cancer cells. J Exp Clin Cancer Res. 2017;36:15. https://doi.org/10.1186/s13046-017-0490-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Qin Y, et al. A novel long non-coding RNA lnc030 maintains breast cancer stem cell stemness by stabilizing SQLE mRNA and increasing cholesterol synthesis. Adv Sci. 2021;8:2002232. https://doi.org/10.1002/advs.202002232.

    Article  CAS  Google Scholar 

  124. Ni W, et al. Targeting cholesterol biosynthesis promotes anti-tumor immunity by inhibiting long noncoding RNA SNHG29-mediated YAP activation. Mol Ther. 2021;29:2995–3010. https://doi.org/10.1016/j.ymthe.2021.05.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Wang H, et al. The lncRNA ZFAS1 regulates lipogenesis in colorectal cancer by binding polyadenylate-binding protein 2 to stabilize SREBP1 mRNA. Mol Ther Nucleic Acids. 2022;27:363–74. https://doi.org/10.1016/j.omtn.2021.12.010.

    Article  CAS  PubMed  Google Scholar 

  126. Jia Y, et al. Long non-coding RNA NEAT1 mediated RPRD1B stability facilitates fatty acid metabolism and lymph node metastasis via c-Jun/c-Fos/SREBP1 axis in gastric cancer. J Exp Clin Cancer Res. 2022;41:287. https://doi.org/10.1186/s13046-022-02449-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Su GH, Xiao Y, Jiang YZ, Shao ZM. Uncovering the molecular subtypes of triple-negative breast cancer with a noninvasive radiomic methodology. Cell Rep Med. 2022;3:100808. https://doi.org/10.1016/j.xcrm.2022.100808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Barisciano G, et al. miR-27a is a master regulator of metabolic reprogramming and chemoresistance in colorectal cancer. Br J Cancer. 2020;122:1354–66. https://doi.org/10.1038/s41416-020-0773-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Li Q, et al. CircACC1 regulates assembly and activation of AMPK complex under metabolic stress. Cell Metab. 2019;30:157-173.e157. https://doi.org/10.1016/j.cmet.2019.05.009.

    Article  CAS  PubMed  Google Scholar 

  130. Balihodzic A, et al. Non-coding RNAs and ferroptosis: potential implications for cancer therapy. Cell Death Differ. 2022;29:1094–106. https://doi.org/10.1038/s41418-022-00998-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Hajibabaie F, Abedpoor N, Mohamadynejad P. Types of cell death from a molecular perspective. Biology. 2023. https://doi.org/10.3390/biology12111426.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Luo Y, et al. Long noncoding RNA LINC01606 protects colon cancer cells from ferroptotic cell death and promotes stemness by SCD1-Wnt/beta-catenin-TFE3 feedback loop signalling. Clin Transl Med. 2022;12:e752. https://doi.org/10.1002/ctm2.752.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Zhang Y, Luo M, Cui X, O’Connell D, Yang Y. Long noncoding RNA NEAT1 promotes ferroptosis by modulating the miR-362-3p/MIOX axis as a ceRNA. Cell Death Differ. 2022;29:1850–63. https://doi.org/10.1038/s41418-022-00970-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Han Y, et al. Long noncoding RNA LINC00239 inhibits ferroptosis in colorectal cancer by binding to Keap1 to stabilize Nrf2. Cell Death Dis. 2022;13:742. https://doi.org/10.1038/s41419-022-05192-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zhang R, et al. Curcumenol triggered ferroptosis in lung cancer cells via lncRNA H19/miR-19b-3p/FTH1 axis. Bioactive Mater. 2022;13:23–36. https://doi.org/10.1016/j.bioactmat.2021.11.013.

    Article  CAS  Google Scholar 

  136. Yang L, et al. Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J Hematol Oncol. 2023;16:59. https://doi.org/10.1186/s13045-023-01453-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Xia L, et al. The cancer metabolic reprogramming and immune response. Mol Cancer. 2021;20:28. https://doi.org/10.1186/s12943-021-01316-8.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Li J, et al. Heat shock factor 1 epigenetically stimulates glutaminase-1-dependent mTOR activation to promote colorectal carcinogenesis. Mol Ther. 2018;26:1828–39. https://doi.org/10.1016/j.ymthe.2018.04.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Liu L, et al. Long non-coding RNA HOTAIR acts as a competing endogenous RNA to promote glioma progression by sponging miR-126-5p. J Cell Physiol. 2018;233:6822–31. https://doi.org/10.1002/jcp.26432.

    Article  CAS  PubMed  Google Scholar 

  140. Bhutia YD, Babu E, Ramachandran S, Ganapathy V. Amino acid transporters in cancer and their relevance to “glutamine addiction”: novel targets for the design of a new class of anticancer drugs. Can Res. 2015;75:1782–8. https://doi.org/10.1158/0008-5472.Can-14-3745.

    Article  CAS  Google Scholar 

  141. Dong J, et al. Epigenetic silencing of microRNA-137 enhances ASCT2 expression and tumor glutamine metabolism. Oncogenesis. 2017;6:e356. https://doi.org/10.1038/oncsis.2017.59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Wang R, et al. LncRNA GIRGL drives CAPRIN1-mediated phase separation to suppress glutaminase-1 translation under glutamine deprivation. Sci Adv. 2021. https://doi.org/10.1126/sciadv.abe5708.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Hao Y, et al. Oncogenic PIK3CA mutations reprogram glutamine metabolism in colorectal cancer. Nat Commun. 2016;7:11971. https://doi.org/10.1038/ncomms11971.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Wang Y, Tang Q, Li M, Jiang S, Wang X. MicroRNA-375 inhibits colorectal cancer growth by targeting PIK3CA. Biochem Biophys Res Commun. 2014;444:199–204. https://doi.org/10.1016/j.bbrc.2014.01.028.

    Article  CAS  PubMed  Google Scholar 

  145. Amelio I, Cutruzzolá F, Antonov A, Agostini M, Melino G. Serine and glycine metabolism in cancer. Trends Biochem Sci. 2014;39:191–8. https://doi.org/10.1016/j.tibs.2014.02.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Zhang G, et al. Energy stress-induced linc01564 activates the serine synthesis pathway and facilitates hepatocellular carcinogenesis. Oncogene. 2021;40:2936–51. https://doi.org/10.1038/s41388-021-01749-x.

    Article  CAS  PubMed  Google Scholar 

  147. Liu X, et al. CircMYH9 drives colorectal cancer growth by regulating serine metabolism and redox homeostasis in a p53-dependent manner. Mol Cancer. 2021;20:114. https://doi.org/10.1186/s12943-021-01412-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Liu Y, et al. Lipid metabolism-related lncRNA SLC25A21-AS1 promotes the progression of oesophageal squamous cell carcinoma by regulating the NPM1/c-Myc axis and SLC25A21 expression. Clin Transl Med. 2022;12:e944. https://doi.org/10.1002/ctm2.944.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Bachmann AS, Geerts D. Polyamine synthesis as a target of MYC oncogenes. J Biol Chem. 2018;293:18757–69. https://doi.org/10.1074/jbc.TM118.003336.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. He W, et al. Targeting ornithine decarboxylase (ODC) inhibits esophageal squamous cell carcinoma progression. NPJ Precis Oncol. 2017;1:13. https://doi.org/10.1038/s41698-017-0014-1.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Meng X, et al. Roles of lncRNA LVBU in regulating urea cycle/polyamine synthesis axis to promote colorectal carcinoma progression. Oncogene. 2022;41:4231–43. https://doi.org/10.1038/s41388-022-02413-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Chen M, et al. Long noncoding RNA LINC01234 promotes hepatocellular carcinoma progression through orchestrating aspartate metabolic reprogramming. Mol Ther. 2022;30:2354–69. https://doi.org/10.1016/j.ymthe.2022.02.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Li Y, et al. HBXIP and LSD1 scaffolded by lncRNA hotair mediate transcriptional activation by c-Myc. Cancer Res. 2016;76:293–304. https://doi.org/10.1158/0008-5472.CAN-14-3607.

    Article  CAS  PubMed  Google Scholar 

  154. Sun Y, et al. LINC01431 promotes histone H4R3 methylation to impede HBV covalently closed circular DNA transcription by stabilizing PRMT1. Adv Sci. 2022;9:e2103135. https://doi.org/10.1002/advs.202103135.

    Article  CAS  Google Scholar 

  155. Meng N, et al. Small protein hidden in lncRNA LOC90024 promotes “cancerous” RNA splicing and tumorigenesis. Adv Sci. 2020;7:1903233. https://doi.org/10.1002/advs.201903233.

    Article  CAS  Google Scholar 

  156. Kaviani E, et al. System biology analysis to develop diagnostic biomarkers, monitoring pathological indexes, and novel therapeutic approaches for immune targeting based on maggot bioactive compounds and polyphenolic cocktails in mice with gastric cancer. Environ Res. 2023;238:117168. https://doi.org/10.1016/j.envres.2023.117168.

    Article  CAS  PubMed  Google Scholar 

  157. Wang X, et al. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Mol Oncol. 2020;14:539–55. https://doi.org/10.1002/1878-0261.12629.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Zheng H, et al. LncRNA XIST/miR-137 axis strengthens chemo-resistance and glycolysis of colorectal cancer cells by hindering transformation from PKM2 to PKM1. Cancer Biomark. 2021;30:395–406. https://doi.org/10.3233/cbm-201740.

    Article  CAS  PubMed  Google Scholar 

  159. Shi H, et al. LncRNA-DANCR interferes with miR-125b-5p/HK2 axis to desensitize colon cancer cells to cisplatin vis activating anaerobic glycolysis. Front Oncol. 2020. https://doi.org/10.3389/fonc.2020.01034.

    Article  PubMed  PubMed Central  Google Scholar 

  160. He W, et al. MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene. 2019;38:4637–54. https://doi.org/10.1038/s41388-019-0747-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Tu MJ, et al. MicroRNA-1291-5p sensitizes pancreatic carcinoma cells to arginine deprivation and chemotherapy through the regulation of arginolysis and glycolysis. Mol Pharmacol. 2020;98:686–94. https://doi.org/10.1124/molpharm.120.000130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Li Y, et al. miR-34a regulates multidrug resistance via positively modulating OAZ2 signaling in colon cancer cells. J Immunol Res. 2018. https://doi.org/10.1155/2018/7498514.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Shi L, et al. LncRNA GLTC targets LDHA for succinylation and enzymatic activity to promote progression and radioiodine resistance in papillary thyroid cancer. Cell Death Differ. 2023;30:1517–32. https://doi.org/10.1038/s41418-023-01157-6.

    Article  CAS  PubMed  Google Scholar 

  164. Guo Y, Wang L, Yang H, Ding N. Knockdown long non-coding RNA HCP5 enhances the radiosensitivity of esophageal carcinoma by modulating AKT signaling activation. Bioengineered. 2022;13:884–93. https://doi.org/10.1080/21655979.2021.2014386.

    Article  CAS  PubMed  Google Scholar 

  165. Lei C, et al. LncRNA DUXAP8 induces breast cancer radioresistance by modulating the PI3K/AKT/mTOR pathway and the EZH2-E-cadherin/RHOB pathway. Cancer Biol Ther. 2022;23:1–13. https://doi.org/10.1080/15384047.2022.2132008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Chen W, et al. Roles of the SNHG7/microRNA-9-5p/DPP4 ceRNA network in the growth and (131)I resistance of thyroid carcinoma cells through PI3K/Akt activation. Oncol Rep. 2021. https://doi.org/10.3892/or.2021.7954.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Zhang J, et al. Docetaxel resistance-derived LINC01085 contributes to the immunotherapy of hormone-independent prostate cancer by activating the STING/MAVS signaling pathway. Cancer Lett. 2022;545:215829. https://doi.org/10.1016/j.canlet.2022.215829.

    Article  CAS  PubMed  Google Scholar 

  168. Song H, et al. Long noncoding RNA CASC11 promotes hepatocarcinogenesis and HCC progression through EIF4A3-mediated E2F1 activation. Clin Transl Med. 2020;10:e220. https://doi.org/10.1002/ctm2.220.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Chen S, et al. Triggering of lymphocytes by CD28, 4–1BB, and PD-1 checkpoints to enhance the immune response capacities. PLoS ONE. 2022. https://doi.org/10.1371/journal.pone.0275777.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Ham IH, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 2019;18:68. https://doi.org/10.1186/s12943-019-0972-8.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Hu Q, et al. Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression. Nat Immunol. 2019;20:835–51. https://doi.org/10.1038/s41590-019-0400-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Han J, et al. MSC-induced lncRNA AGAP2-AS1 promotes stemness and trastuzumab resistance through regulating CPT1 expression and fatty acid oxidation in breast cancer. Oncogene. 2021;40:833–47. https://doi.org/10.1038/s41388-020-01574-8.

    Article  CAS  PubMed  Google Scholar 

  173. Ling Y, et al. circCDYL2 promotes trastuzumab resistance via sustaining HER2 downstream signaling in breast cancer. Mol Cancer. 2022;21:8. https://doi.org/10.1186/s12943-021-01476-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Zheng J, et al. CircFOXK2 promotes hepatocellular carcinoma progression and leads to a poor clinical prognosis via regulating the Warburg effect. J Exp Clin Cancer Res. 2023;42:63. https://doi.org/10.1186/s13046-023-02624-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Huo N, et al. STAT3/LINC00671 axis regulates papillary thyroid tumor growth and metastasis via LDHA-mediated glycolysis. Cell Death Dis. 2021;12:799. https://doi.org/10.1038/s41419-021-04081-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Han J, Chen X, Wang J, Liu B. Glycolysis-related lncRNA TMEM105 upregulates LDHA to facilitate breast cancer liver metastasis via sponging miR-1208. Cell Death Dis. 2023;14:80. https://doi.org/10.1038/s41419-023-05628-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Wu H, et al. Kaempferol reverses aerobic glycolysis via miR-339-5p-mediated PKM alternative splicing in colon cancer cells. J Agric Food Chem. 2021;69:3060–8. https://doi.org/10.1021/acs.jafc.0c07640.

    Article  CAS  PubMed  Google Scholar 

  178. Shi H, Li K, Feng J, Zhang X. Overexpression of long non-coding RNA urothelial carcinoma associated 1 causes paclitaxel (Taxol) resistance in colorectal cancer cells by promoting glycolysis. J Chemother. 2021;33:409–19. https://doi.org/10.1080/1120009x.2021.1906032.

    Article  CAS  PubMed  Google Scholar 

  179. Gao SJ, Ren SN, Liu YT, Yan HW, Chen XB. Targeting EGFR sensitizes 5-Fu-resistant colon cancer cells through modification of the lncRNA-FGD5-AS1-miR-330-3p-Hexokinase 2 axis. Mol Ther Oncol. 2021;23:14–25. https://doi.org/10.1016/j.omto.2021.06.012.

    Article  CAS  Google Scholar 

  180. Liu J, et al. Long noncoding RNA AGPG regulates PFKFB3-mediated tumor glycolytic reprogramming. Nat Commun. 2020;11:1507. https://doi.org/10.1038/s41467-020-15112-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Chen F, et al. Extracellular vesicle-packaged HIF-1alpha-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol. 2019;21:498–510. https://doi.org/10.1038/s41556-019-0299-0.

    Article  CAS  PubMed  Google Scholar 

  182. Hajibabaie F, et al. The molecular perspective on the melanoma and genome engineering of T-cells in targeting therapy. Environ Res. 2023;237:116980. https://doi.org/10.1016/j.envres.2023.116980.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported in part by grants from the following sources: the National Natural Science Foundation of China (82203233, 82202966, 82173142, 81972636), the Natural Science Foundation of Hunan Province (2023JJ60469, 2023JJ40413, 2023JJ30372, 2023JJ30375, 2022JJ80078, 2020JJ5336), the science and technology innovation Program of Hunan Province (2023RC1073, 2023RC3199), the Research Project of Health Commission of Hunan Province (202203034978, 202109031837, 20201020), Hunan Provincial Science and Technology Department (2020TP1018), Ascend Foundation of National cancer center (NCC201909B06), and by Hunan Cancer Hospital Climb Plan (ZX2020001-3, YF2020002).

Author information

Authors and Affiliations

Authors

Contributions

SL, MP, ST, LO, JL, XL, JW, NW, XJ and QP collected the related papers and drafted the manuscript YZ, and QL revised and finalized the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yujuan Zhou or Qianjin Liao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, S., Peng, M., Tan, S. et al. The roles and molecular mechanisms of non-coding RNA in cancer metabolic reprogramming. Cancer Cell Int 24, 37 (2024). https://doi.org/10.1186/s12935-023-03186-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-023-03186-0

Keywords