International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Digestive system neoplasms are the most common malignant tumor in China, mainly including esophageal squamous cell carcinoma (ESCC), gastric cancer (GC), colorectal cancer (CRC), hepatocellular carcinoma (HCC) and pancreatic cancer (PC) (1,2). Notably, CRC is the second leading cause of global cancer mortality, and GC ranks fourth. Furthermore, the incidence rates of both cancers remain persistently high. A pivotal reason for the high mortality is metastasis, which accounts for ~90% of cancer deaths. The metastatic process entails essential steps such as epithelial-mesenchymal transition (EMT), tumor invasion, intravasation, extravasation and mesenchymal-epithelial transition (MET), culminating in the establishment of distant metastases (Fig. 1). In the present review, ‘invasion’ specifically refers to the ability of cells to degrade and penetrate the extracellular matrix or basement membrane, emphasizing its destructive nature; ‘migration’ describes the motility of cells such as tumor cells; and ‘metastasis’ denotes the multistep process by which tumor cells disseminate from the primary site to distant organs and form secondary tumors (3,4).
Emerging evidence indicates that metabolic reprogramming of tumor cells serves as a critical driver of tumor metastasis. Malignant tumor cells alter metabolic reprogramming through the modulation of conventional activities of key enzymes, as well as the regulation of their non-canonical roles (5–7). Meanwhile, several metabolic pathways have been reported to be involved in metabolic reprogramming to meet increased bioenergetic and biosynthetic needs to support cancer cell proliferation, invasion and metastasis (8). With the growing identification of aberrantly expressed non-coding (nc)RNAs in tumors, these regulatory molecules have been reported to induce extensive metabolic crosstalk through targeting rate-limiting enzymes, ultimately forming sophisticated regulatory networks. The present review summarizes abnormal substance metabolism in digestive system tumor metastasis and the related metabolic pathway networks.
A hallmark of cancer is aberrant metabolism. Cells often metabolize glucose, and cancer cells preferentially use glycolysis over mitochondrial oxidative phosphorylation to create glucose-dependent ATP and glycoferment intermediates for macromolecular biosynthesis, even in conditions where oxygen supply is sufficient. This is referred to as the Warburg effect (9–11). Important players in glycolytic pathways include pyruvate kinase (PK), phosphofructokinase (PFK) and HK. Moreover, research has reported that a number of coding RNAs and ncRNAs are notable regulators of the metabolic enzymes and signaling pathways involved in the metabolism of glucose in cancer cells (12,13) (Table I and Fig. 2).
HK phosphorylates glucose to G6P in an ATP-dependent manner, catalyzing the first irreversible step in the glycolytic pathway. The four primary isoenzymes, HK1, HK2, HK3 and HK4, are encoded by several genes (12). The primary isoenzyme, HK2, is engaged in the first and rate-limiting phases of glycolysis, which transforms glucose into glucose-6-phosphate. Several cancer types show elevated levels of this compound due to its ability to enhance glucose uptake in cells and the Warburg effect (13,14). Guo et al (15) identified a putative competing endogenous (ce)RNA regulatory network in which the lysyl oxidase like 1 (LOXL1)/microRNA (miR)-1224-5p/miR-761/HK2 axis was shown to promote cell proliferation and metastasis by regulating aerobic glucose metabolism in CRC HCT-116 and SW480 cells, wherein the long non-coding (lnc)RNA LOXL1 modulated HK2 expression through competitively binding to endogenous miR-1224-5p and miR-761. Similarly, lncRNA MIR210HG was reported to be abnormally upregulated in PC, and promote lactate accumulation through the miR-125b-5p/HK2/PKM2 axis, ultimately affecting malignant phenotypes, including proliferation, invasion and migration in PC PANC-1 and MIA PaCa-2 cells (16). Additionally, Chen et al (17) reported that sponging of miR-125b reduced the HK2-mediated Warburg effect in HCC Hep3B cells, and sponging of miR-125b downregulated hsa_circ_0001806 expression, thereby suppressing the HK2-mediated Warburg effect, primarily through inhibiting lactate accumulation and ATP production. This ultimately impeded tumor proliferation and migration. Several studies have reported that circPRMT5, hsa_circ_0045932 and circRPS19 actively regulate the expression of HK2 via sponge miRNAs, thereby accelerating glycolysis and promoting tumor development (18–20). Another isoenzyme of hexokinase, HK1, is reported to be positively regulated by the lncRNA ARSR. This lncRNA binds to miR-34a-5p through a sponge adsorption mechanism, thereby promoting glucose uptake, lactate and ATP production in CRC Caco-2 cells, and ultimately enhancing the invasion and migration capabilities of tumor cells (21).
The rate-limiting enzyme PK catalyzes the last stage of glycolysis, which is the conversion of phosphoenol pyruvate to pyruvate (22). There are several subtypes of mammalian PK, of which PKM2 is directly involved in cancer aerobic glycolysis. Although mitochondrial oxidative phosphorylation generates more net energy than aerobic glycolysis in tumors, cancer cells use the Warburg effect to digest glucose more quickly in response to higher energy demands. PKM2 is thought to be a crucial metabolic enzyme for the Warburg effect in cancer cells, and the overexpression of this enzyme is primarily associated with an increased use of glucose and altered redox balance in cells (23). Increasing evidence points to the possible involvement of PKM2 in tumor metastasis (24–26). PKM2 proteins are bound by the lncRNA FEZF1-antisense RNA (AS)1 (FEZF1-AS1), which enhances their stability and raises the amounts of PKM2 in the cytoplasm and nucleus. Whilst FEZF1-AS1 regulates PKM2/STAT3 signaling and glycolysis to increase the proliferation and migration of CRC LoVo and HCT116 cells, an increase in cytosolic PKM2 stimulates PK activity and lactate generation (27). Notably, under hypoxic conditions, miR-338-3p interacts with circRNA MAT2B, thereby upregulating PKM2 expression levels and enhancing glycolysis in HCC Huh7 and HepG2 cells. The ATP generated from this metabolic reprogramming directly fuels cytoskeletal remodeling and membrane fluidity, whilst lactate accumulation contributes to creating an acidic microenvironment conducive to invasion, collectively promoting tumor progression (28).
One of the major glycolytic enzymes, lactate dehydrogenase A (LDHA) is highly associated with thyroid cancer, CRC and pancreatic ductal adenocarcinoma (PDAC) malignancies due to its aberrant expression and overexpression (29–31). Regenerating islet-derived protein 1-α (REG1α), for instance, was reported to be upregulated in CRC serum and tissues. By enhancing glycolysis mediated by the β-catenin/MYC axis, REG1α promoted invasion in CRC DLD-1 and HCT-116 cells, and lung metastasis in BALB/c nude mouse models. Furthermore, the Wnt/β-catenin/MYC axis or glycolysis pathway could be silenced to successfully reverse the malignant phenotype controlled by REG1α. Furthermore, REG1α expression was moderated by methyltransferase-like 3 (METTL3). The METTL3/REG1α/β-catenin/MYC axis in CRC suggests that REG1α may serve as a novel biomarker and a possible target for treatment in patients with CRC. REG1α drives glycolysis to provide both the energy and carbon sources required for the synthesis and activation of matrix metalloproteinases, which are essential for extracellular matrix degradation during tumor cell invasion (30). Nucleolar and spindle associated protein 1 (NUSAP1) was markedly overexpressed in PDAC, and in several PDAC cell lines, NUSAP1 expression was consistent with that of LDHA. Additionally, NUSAP1 facilitated PDAC metastasis and LDHA-mediated glycolysis in BALB/c nude mouse models (31). This suggests that NUSAP1 promotes the formation of distant metastases by regulating lactate production and consequently influencing the pH of the TME. Phosphoglycerate kinase 1 (PGK1) and 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB), are key components of the glycolytic pathway. Furthermore, PFKFB has four isoenzymes (PFKFB1, PFKFB2, PFKFB3 and PFKFB4), and studies have reported that PFKFB3 and PFKFB4 are involved in EMT and associated with tumor metastasis (32). Moreover, linc01572 modulated HCC Huh7 and SNU-449 cells invasion and migration by increasing PFKFB4 levels by sponging miR-195-5p and subsequently enhancing glycolysis and PI3K-AKT signaling activation (33).
The Warburg effect begins with the transfer of glucose from the extracellular milieu. Using simple diffusion, the transport capacity of glucose is determined by its hydrophilicity. Consequently, certain membrane transporters are needed for this process. In this mechanism, the GLUT family is crucial (34). Out of the 14 GLUT family members, GLUT1 is most frequently expressed, which is also recognized as the basal switch for glycolysis in tumor cells (35). Research has revealed that low survival rates in human cancer are associated with elevated GLUT1 expression, potentially due to its association with the presence of metastases (36). The promotion of cancer cell proliferation and metastatic behavior is closely associated with GLUT1 overexpression (37). Shang et al (38) reported that lncRNA SLC2A1-AS1 is downregulated in liver cancer tissues. This lncRNA was shown to inhibit aerobic glycofermentation and the progression of HCC MHCC97-H and Huh7 cells by inactivating GLUT1, which is encoded by the opposite-strand-encoding gene SLC2A1. Meanwhile, lncRNA SLC2A1-AS1 enhanced GLUT1 expression through sponging miR-378a-3p, thereby promoting aerobic glycolysis in ESCC EC9706, TE1 and KYSE180 cells, and ultimately facilitating tumor cell invasion and migration through the EMT pathway (39). Furthermore, FOXD1 was upregulated in PC, and it activated SLC2A1 and the lncRNA HOXA11-AS by sponging miR-148b. Reduced FOXD1 inhibited MIA PaCa-2 and PANC-1 PC cells invasion, migration and aerobic glycolysis. Furthermore, a positive correlation was observed between FOXD1 and GLUT1, suggesting their synergistic role in driving oncogenesis (40). According to recent research, GLUT3 stimulates the EMT process via the TGF-β/JNK/ATF2 signaling pathway, suggesting that this system may be a target for the treatment of metastatic CRC (41). Moreover, by controlling aerobic glycolysis to stimulate GC tumor growth and migration, the oncogenic role of GLUT12 may be targeted by the let-7a-5p/GLUT12 pathway to amplify the anticancer properties of everolimus, suggesting a promising and viable therapeutic strategy (42).
Solid tumors are characterized by hypoxia, which is a diminished oxygen supply that promotes tumor growth. Hypoxic conditions induce molecular responses in both normal and tumor cells, driving the activation of the key transcription factor HIF (43). As a crucial regulator of oxygen balance, HIF-1 is a heterodimer transcription factor made up of stable HIF-1β subunits and unstable HIF-1α subunits (44). HIF-1α is a transcription factor that is frequently activated by the hypoxic milieu found in tumors and is intimately associated with the growth, metastasis and angiogenesis of tumors (45). It has been established that lncRNA SNHG11 stimulates HIF-1α downstream targets to enhance CRC HCT-116 and RKO cell invasion and migration (46). It has also been reported that upregulation of TMEM161B-AS1 promoted the expression of HIF1AN via absorbing miR-23a-3p, inhibiting Eca109 and KYSE30 proliferation, invasion and glycolysis, further inducing the downregulation of glycoenzyme-associated proteins, and ultimately leading to the inhibition of ESCC progression (47). Furthermore, research has reported that circRNF13 expression increased in PC and was associated with a low rate of survival. The signaling pathway activated by HIF-1α and RNF13 used the miR-654/PDK3 axis to increase PC cell invasion in vitro and metastasis in BALB/c nude mouse lung and liver metastasis models (48). By enhancing PDK3 activity, this axis strengthens glycolytic metabolism, thereby providing the energy required for the formation of invaders and the activation of proteases, which mediate the degradation of extracellular matrix during distant local invasion and extravasation processes. Exosome release was stimulated by hypoxia, and miR-301 expression was raised. Exosomes may carry enhanced miR-301a-3p from one GC cell to another. The transferred miR-301a-3p subsequently helped to block HIF-1α degradation by focusing on prolyl hydroxylase domain-containing protein 3, which could hydroxylate HIF-1α subunits and ubiquitinate their destruction. HIF-1α and miR-301a-3p created a positive feedback loop that promoted GC invasion, migration, proliferation and EMT in BALB/c nude mouse models of lung metastasis (49).
An important glucose catabolic route is the PPP. The PPP involves numerous enzymes and is essential in controlling the proliferation of cancer cells (50).
One of the essential enzymes in the PPP oxidation branch, 6PGD is involved in redox and nucleotide biosynthesis (51). In several studies, elevated G6PD levels have been reported to enhance the cancer progression in several tumor types (52–54). Through the modulation of important enzymes, ncRNAs perform their roles in PPP regulation. In HCC MHCC97H and HCCLM3 cells, G6PD promotes migration and invasion by activating the STAT3 pathways, which in turn causes EMT (52). circ_0003215 inhibits invasion and migration in CRC SW480 and HT29 cells by suppressing the PPP through DLG4-mediated, K48-linked ubiquitination and degradation of G6PD (54). Additionally, circNOLC1 interacts with AZGP1 to activate the mTOR/SREBP1 signaling pathway, or functions as a sponge for miR-212-5p to upregulate c-Met expression. Collectively, these mechanisms induce G6PD to enhance the PPP in CRC HCT116 and LoVo cells, promoting liver metastasis in CRC (55). Furthermore, it has been reported that NeuroD1 positively regulates G6PD in CRC and alters tumor cell metabolism by stimulating the PPP, therapy enhancing CRC tumorigenesis (56).
In the non-oxidized phase of PPP, TKT is an essential enzyme that helps to maintain the levels of ribose 5-phosphate. The transketase family includes three human genes, TKT and TKT-like genes 1 and 2 (57). TKT has been reported to be upregulated in HCC and CRC (58,59), and it has been demonstrated to have a positive association with HCC metastasis and proliferation. Previous research reported that TKT controlled metastasis in CRC via attaching to GRP78, which changed AKT phosphorylation to influence cell glycolysis (59). In the meantime, ncRNAs took part in networks connected to TKT. Through its interactions with TKT and STAT3, the lncRNA TSLNC8 inactivated the IL6/STAT3 signaling pathway, suppressing STAT3 phosphorylation and transcriptional activity in HCC Huh-7 cells. This in turn prevented the growth of tumors (60). Zeng et al (61) reported that downregulation of S100A11 suppressed the malignant progression of PDAC and inhibited the PPP via TKT expression.
The TCA cycle satisfies the needs of cells for bioenergetics, biosynthesis and redox equilibrium, and is a key route for oxidative phosphorylation. Research indicates that certain cancer cells, particularly those with dysregulated oncogene and tumor suppressor gene expression, may heavily depend on the TCA cycle to produce energy and synthesize macromolecules, despite the earlier hypothesis that cancer cells primarily used aerobic glycolysis and avoided the TCA cycle (62).
There are four isoenzymes that make up pyruvate dehydrogenase kinases (PDKs). The pyruvate dehydrogenase complex is phosphorylated and rendered inactive by PDKs, preventing mitochondria from oxidatively metabolizing pyruvate. This procedure may stimulate anabolic pathways, which in turn may stimulate the growth of cancer cells. By raising pyruvate dehydrogenase (PDH) activity and thus increasing ROS generation, inhibition of PDK causes cell death (63).
A gatekeeper enzyme called PDK1 is involved in the growth, survival and angiogenesis of tumors in cancer. PDK1 is a regulator of PDH, a key rate-limiting enzyme of cellular glycolysis, and has been widely confirmed to be involved in the regulation of cancer progression (64). Research has reported that circ_0000284 serves an oncogene role in intrahepatic cholangiocarcinoma (ICC), which facilitates ICC HCCC-9810 and HUCCT1 cell growth and metastasis through sponging miR-152-3p to increase PDK1 expression (65). According to another study, by sponging miR-599, lncRNA PTPRG-AS1 upregulated PDK1 expression, thereby promoting migration and proliferation in ESCC TE-8 and KYSE-150 cells (66). In GC SGC7901 and MGC803 cells, to restore the activity of PDH, the gatekeeping enzyme that catalyzes the decarboxylation of pyruvate to create acetyl-CoA, miR-422a was reported to suppress PDK2. This process reduced NADPH generation, thereby compromising the ability of the cell to counteract oxidative damage incurred during migration (67).
Lipids are taken in by mammalian cells by two different mechanisms: Uptake and de novo synthesis. Furthermore, fat accumulation in cells may encourage the growth, colonization and ability of tumor cells to metastasize. Tumor cells go through several stages of metastasis, requiring modifications to their metabolism and structure related to lipids (68). Moreover, there is growing evidence that lipid metabolism is essentially reprogrammed in cancer (69,70) (Table II and Fig. 2).
A class of intracellular lipid chaperones known as FABPs binds to long-chain fatty acids and related lipids to help move them into different parts of the cell, including the nucleus (71). Humans have been found to exhibit 10 distinct FABP subtypes. FABP5, a diminutive constituent of the cytoplasmic FABP family, exhibits a strong propensity for binding to long-chain FAs. Research suggests that FABP5, through triggering EMT and controlling angiogenic responses, serves a critical role in the invasion, metastasis and development of cancer (72,73). In addition, FABP1 is overexpressed in the cytoplasm of HCC cells (74). However, IL-6 has been reported to increase the expression of miR-603 and then suppresses the production of FABP1, which promotes lipid metabolism and the manufacture of related proteins, ultimately raising the levels of cellular oxidative stress and causing HCC Huh-7 cells migration and invasion (75).
SREBPs are the most important transcription factors for regulating lipid homeostasis. Previous studies have reported that SREBP is highly upregulated and promotes tumor growth in several cancers (70). The lncRNA ZFAS1 has been reported to rewire fat metabolism to accelerate the invasion of CRC SW480 and RKO cells by binding to PABP2 and promoting the interaction between PABP2 and SREBP1 (76). Moreover, the lncRNA SNHG16 was reported to promote invasion and migration by directly controlling the miR-195/SREBP2 axis, which in turn sped up the development of PC PANC-1 and AsPC-1 cells. By increasing cholesterol synthesis, SREBP2 enhances plasma membrane fluidity, which promotes deformability and motility for cell migration. Concurrently, it supplies the essential lipid components required by metastatic cells to preserve membrane integrity (77). Furthermore, through mRNA splicing and transcription, lncRNA MALAT1 stimulates the expression of several genes in AMPK signaling and unsaturated fatty acid metabolism pathways, leading to the upregulation of glucose uptake and adipogenesis, which indicates the function of MALAT1 in tumor cell invasion and migration (78).
Increased expression of the SCD1 enzyme has been reported to accelerate the development of several malignancies. SCD1 is involved in the synthesis of monounsaturated fatty acids, including oleic acid (79). The orthotopic CRC model demonstrated that SULT2B1 promotes metastatic progression of colorectal cancer. Furthermore, experiments in HCT116 and SW480 cells revealed that SULT2B1 directly interacts with SCD to enhance lipid metabolism, thereby facilitating the metastasis of CRC (80). In addition, low expression of miR-4310 is associated with a poor prognosis. By inhibiting SCD1 and FASN-mediated lipid synthesis, miR-4310 has been reported to inhibit HCC cell migration and invasion in vitro and HCC tumor growth and metastasis in vivo (81). Furthermore, Ly1 antibody reactive (LYAR) and fascin actin-bundling protein 1 (FSCN1) has been reported to participate in CRC progression and promote cell metastasis, and LYAR partly regulates FSCN1 expression in CRC. Additionally, FSCN1 is associated with lipid metabolism, and FSCN1 silencing reduces the expression levels of FASN and SCD1 in CRC progression (82).
Serum deprivation-response protein (SDPR) has been reported to participate in TGF-β-induced GC metastasis. It has been shown that overexpression of SDPR inhibits TGF-β-induced GC MKN45 and MGC803 cells invasion and migration by suppressing ERK and enhancing the expression of peroxisome proliferator-activated receptor α, thereby upregulating the expression of carnitine palmitoyltransferase 1A (CPT1A), promoting fatty acid oxidation (FAO) and inducing GC metastasis. By driving FAO, CPT1A supports the TCA cycle function through acetyl-CoA production, which in turn furnishes the persistent energy required for cell migration (83). Zhao et al (84) reported that hexokinase domain containing 1 (HKDC1) expression was upregulated in GC. Additionally, lipid metabolism serves a critical role in the role of PRKDC as a downstream effector of HKDC1-mediated GC carcinogenesis. HKDC1 can mechanically regulate the well-known oncoprotein G3BP stress granule assembly factor 1 (G3BP1), and HKDC1 and G3BP1 collaborate to support PRKDC stability. The HKDC1/G3BP1-PRKDC axis reprograms lipid metabolism to promote GC metastasis and chemoresistance (84).
Glutamine is mostly utilized for the synthesis of energy and is regarded as a non-essential amino acid. However, when certain cancer cells proliferate quickly, when there is stress, or when proliferating cells grow quickly, glutamine becomes a conditional necessary amino acid (85,86). SLC1A5 is a sodium-coupled transporter of alanine, serine, cysteine and glutamine. It is often referred to as alanine-serine-cysteine transporter 2. The expression of circ_0001273 is upregulated in esophageal cancer (EC) tissues and cell lines. As circ_0001273 regulates miR-622 and downstream SLC1A5, it partially explains why silencing EC TE1 and ECA109 cells was reported to reduce their survival, proliferation, migration and rate of glutamine metabolism (87). Moreover, circ_0001093 has been reported to function as ceRNA by sponging miR-579-3p, raising the expression of glutaminase, enhancing glutamine metabolism and advancing malignancy in ESCC (88). Building on the established role of glutamic-oxaloacetic transaminase 1 (GOT1) in glutamate metabolism (89), further research demonstrates that silencing circ_MBOAT2 inhibits glutamine catabolism and progression in PC PANC-1 and SW1990 cells via the circ_MBOAT2/miR-433-3p/GOT1 axis (90). Furthermore, HIF-2α is markedly upregulated in PDAC Panc-1 and Capan-2 cells and promotes glutamine metabolism by targeting GOT1 via activation of the PI3K/mTOR complex 2 pathways (91). Low GOT2 expression is also associated with glutamine metabolic reprogramming to glutathione synthesis, and promotes HCC progression by activating the PI3K/AKT/mTOR pathway (92) (Table III).
Mitochondria influence tumorigenesis, and as the primary source of ATP, mitochondria provide building blocks for anabolism through cellular repair mechanisms, with their pivotal role in regulated cell death signaling crucial for tumor dissemination (93). For instance, transmembrane guanylate cyclase natriuretic peptide receptor 1 facilitates GC lymph node metastasis by activating lipid droplet lipolysis and enhancing mitochondrial oxidative phosphorylation (OXPHOS) (94). Moreover, organic cation transporter novel type 2 augments cancer stem-like traits in HCC through increased fatty acid β-oxidation and OXPHOS activation (95) (Fig. 3).
The OXPHOS metabolic pathway fulfills two critical functions in driving tumor cell metastasis: i) Providing bioenergetic demands in the form of ATP; and ii) Extracting carbon from glucose for macromolecule synthesis, serving as a central hub integrating catabolic and anabolic processes. Enzymes of the TCA cycle in the mitochondrial matrix and transmembrane protein complexes of the electron transport chain (ETC) constitute the core machinery for this process. Within the mitochondrial electron transport system, ETC complexes I–IV accept electrons from TCA cycle-derived NADH and FADH2. Subsequent proton reflux through Complex V (ATP synthase) into the mitochondrial matrix generates ATP (96). Certain tumors exhibit high dependency on OXPHOS-derived ATP, which can be identified through specific genetic alterations (97). Metastasis-associated antigen 1 has been identified as a novel regulator of ATP synthase via its interaction with the ATP synthase F1 subunit α, promoting colon cancer liver metastasis through mitochondrial bioenergetic reprogramming and enhanced OXPHOS (98). Sorting nexin 17 facilitates HCC proliferation and metastasis by interacting with STAT3, thereby activating the STAT3/c-Myc signaling pathway to augment OXPHOS (99).
CPT1A is a rate-limiting enzyme localized on the mitochondrial outer membrane, and catalyzes the rate-limiting step of FAO to generate ATP essential for tumor cell proliferation and metastasis. CPT1A-dependent FAO represents a vital metabolic pathway in malignancies. Elevated CPT1A expression in CRC (100) and PDAC (101) activates FAO and enhances metastatic potential. Furthermore, ALKBH5 promotes CRC progression by mediating CPT1A upregulation through m6A demethylation, consequently facilitating M2 macrophage polarization (102).
ROS exhibit a dual role in oncogenesis: At low-to-moderate levels, mtROS function as crucial signaling molecules that activate several protumor signaling pathways through oxidative modification (103). For instance, low-level mtROS stabilizes HIF-1α to enhance glycolysis and facilitate metastasis (104), whereas high-level mtROS conversely activates the SIRT3/PGC-1α axis to promote OXPHOS (105). Furthermore, fatty acid β-oxidation inherently generates ROS as metabolic byproducts. Although FAO is recognized as a tumor-promoting process, the metabolic crosstalk triggered by FAO-derived ROS requires further mechanistic exploration. However, when mtROS accumulation becomes excessive and surpasses the cellular antioxidant defense capacity, it induces severe oxidative stress, triggering cell death, necrosis and ferroptosis, resulting in tumor-suppressive effects (106). For example, CST1 promotes gastric cancer metastasis by recruiting OTUB1 to alleviate GPX4 ubiquitination, thereby enhancing GPX4 protein stability, reducing ROS accumulation, and consequently inhibiting ferroptosis (107). Therefore, strategies aimed at leveraging ROS to block tumor metastasis have emerged as a promising therapeutic approach in oncology.
In recent years, metabolism in tumor metastasis has attracted more attention. However, cancer cell metabolism is a complex matter. The present review discusses, primarily based on evidence from in vitro studies, several metabolic alterations commonly found in cancer cells, including glycolysis, FAO, amino acid metabolism and alterations in mitochondria. This review systematically synthesizes evidence on how key genes target metabolic enzymes or related signaling pathways to drive tumor invasion and metastasis. It aims to establish the correlation between tumor metastasis and cellular metabolism, and to identify suitable targets at the intersection of metabolic pathways to selectively disrupt metabolic networks and prevent tumor metastasis (108,109). For instance, 2-deoxy-d-glucose can inhibit glycolysis and induce cell death (110). However, monotherapy targeting tumor progression still requires further exploration.
Digestive system neoplasms metastasis is one of the leading causes of tumor-related mortality worldwide. The present review identified common features in metabolic reprogramming across gastrointestinal cancers. Beyond the classical Warburg effect, metabolic alterations such as enhanced fatty acid β-oxidation, elevated mitochondrial oxidative phosphorylation, and increased ROS levels all contribute to varying degrees to tumor progression. Furthermore, during the invasion or migration of digestive system tumors, the expression levels of certain key metabolic enzymes are upregulated; for instance, HK expression is increased in HCC (75), colorectal cancer (76), PC (77) and GC (80). However, metabolic specificity also exists among different gastrointestinal tumors, primarily stemming from differences in tissue origin and the TME. For example, enhanced bile acid metabolism promotes the progression of liver cancer: Treatment with T-CDCA was reported to markedly increase the proliferative capacity of HCC HepG2 cells, and downregulate the expression of the tumor suppressor protein CEBPα in HCC (111). Additionally, metabolic dysregulation mediated by gut microbiota serves a crucial role in CRC, involving species such as Streptococcus bovis, Helicobacter pylori and Escherichia coli (112).
Targeting a single metabolic pathway presents notable challenges, whereas a combination of therapies simultaneously targeting multiple tumor metabolites or metabolic enzymes may enhance therapeutic efficacy. Studies have demonstrated that mitochondria generate ATP through multiple pathways, serving as the primary energy source for tumor metastasis. Precise targeting of cancer-specific mitochondria can impair their capacity for de-differentiation, proliferation and metastasis, thereby contributing to improved treatment outcomes and overall prognosis (113). As a result, the individualized role of mitochondria in cancer should receive more attention. The tumor metabolic microenvironment in vivo is complex, and the present review summarizes only a part of the complex events of cancer development. Therefore, researching the function that mitochondrial metabolism serves in the TME can lead to novel therapeutic approaches for cancer, including targeted medication development.
Although preliminary progress has been made in research on tumor metabolic reprogramming, notable knowledge gaps and insufficient investigation remain regarding the role of metabolic crosstalk in tumor metastasis, necessitating further in-depth exploration: i) Most current studies rely on in vitro cell models, which fail to fully recapitulate the complexity and dynamic nature of the TME in vivo. Consequently, conclusions derived from in vitro experiments exhibit certain limitations; ii) tumor metabolism demonstrates high heterogeneity, not only between primary and metastatic sites but also within different regions of a single tumor. To date, systematic studies on metabolic dynamics are still lacking; and iii) although the present review summarizes several metabolic enzymes and metabolites that regulate tumor cell metastasis, the understanding of transcriptome-genome-metabolome crosstalk patterns continues to evolve. Elucidating the mechanisms of metabolic crosstalk in tumor cells will provide new opportunities for effectively combating cancer.
Not applicable.
The present project was supported by the National Natural Science Foundation of China (grant no. 32401139).
Not applicable.
JL conceived, organized and wrote the manuscript. WZ revised the manuscript for important intellectual content. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023.PubMed/NCBI | |
|
Cao M, Li H, Sun D and Chen W: Cancer burden of major cancers in China: A need for sustainable actions. Cancer Commun (Lond). 40:205–210. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chaffer CL and Weinberg RA: A perspective on cancer cell metastasis. Science. 331:1559–1564. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Clark AG and Vignjevic DM: Modes of cancer cell invasion and the role of the microenvironment. Curr Opin Cell Biol. 36:13–22. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Xu D, Shao F, Bian X, Meng Y, Liang T and Lu Z: The evolving landscape of noncanonical functions of metabolic enzymes in cancer and other pathologies. Cell Metab. 33:33–50. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pan C, Li B and Simon MC: Moonlighting functions of metabolic enzymes and metabolites in cancer. Mol Cell. 81:3760–3774. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lv L and Lei Q: Proteins moonlighting in tumor metabolism and epigenetics. Front Med. 15:383–403. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Martínez-Reyes I and Chandel NS: Cancer metabolism: Looking forward. Nat Rev Cancer. 21:669–680. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lin YH, Wu MH, Huang YH, Yeh CT, Cheng ML, Chi HC, Tsai CY, Chung IH, Chen CY and Lin KH: Taurine up-regulated gene 1 functions as a master regulator to coordinate glycolysis and metastasis in hepatocellular carcinoma. Hepatology. 67:188–203. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Warburg O: On the origin of cancer cells. Science. 123:309–314. 1956. View Article : Google Scholar : PubMed/NCBI | |
|
He J, Li F, Zhou Y, Hou X, Liu S, Li X, Zhang Y, Jing X and Yang L: LncRNA XLOC_006390 promotes pancreatic carcinogenesis and glutamate metabolism by stabilizing c-Myc. Cancer Lett. 469:419–428. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Robey RB and Hay N: Is Akt the ‘Warburg kinase’?-Akt-energy metabolism interactions and oncogenesis. Semin Cancer Biol. 19:25–31. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Xu F, Yan JJ, Gan Y, Chang Y, Wang HL, He XX and Zhao Q: miR-885-5p negatively regulates warburg effect by silencing hexokinase 2 in liver cancer. Mol Ther Nucleic Acids. 18:308–319. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yin D, Hua L, Wang J, Liu Y and Li X: Long Non-Coding RNA DUXAP8 facilitates cell viability, migration, and glycolysis in non-small-cell lung cancer via regulating HK2 and LDHA by Inhibition of miR-409-3p. Onco Targets Ther. 13:7111–7123. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Guo T, Peng S, Liu D and Li Y: Long Non-coding RNA LOXL1-AS1 facilitates colorectal cancer progression via regulating miR-1224-5p/miR-761/HK2 axis. Biochem Genet. 60:2416–2433. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu T, Li G, Wang C, Gong G, Wang L, Li C, Chen Y and Wang X: MIR210HG regulates glycolysis, cell proliferation, and metastasis of pancreatic cancer cells through miR-125b-5p/HK2/PKM2 axis. RNA Biol. 18:2513–2530. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, She P, Wang C, Shi L, Zhang T, Wang Y, Li H, Qian L and Li M: Hsa_circ_0001806 promotes glycolysis and cell progression in hepatocellular carcinoma through miR-125b/HK2. J Clin Lab Anal. 35:e239912021. View Article : Google Scholar : PubMed/NCBI | |
|
Ding Z, Guo L, Deng Z and Li P: Circ-PRMT5 enhances the proliferation, migration and glycolysis of hepatoma cells by targeting miR-188-5p/HK2 axis. Ann Hepatol. 19:269–279. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hong F, Deng Z, Tie R and Yang S: Hsa_circ_0045932 regulates the progression of colorectal cancer by regulating HK2 through sponging miR-873-5p. J Clin Lab Anal. 36:e246412022. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng X, Shao J, Qian J and Liu S: circRPS19 affects HK2-mediated aerobic glycolysis and cell viability via the miR-125a-5p/USP7 pathway in gastric cancer. Int J Oncol. 63:982023. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Zhu K, Liu L, Gu J, Niu H and Guo J: lncARSR sponges miR-34a-5p to promote colorectal cancer invasion and metastasis via hexokinase-1-mediated glycolysis. Cancer Sci. 111:3938–3952. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang W, Xia Y, Hawke D, Li X, Liang J, Xing D, Aldape K, Hunter T, Alfred Yung WK and Lu Z: PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell. 150:685–696. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Alquraishi M, Puckett DL, Alani DS, Humidat AS, Frankel VD, Donohoe DR, Whelan J and Bettaieb A: Pyruvate kinase M2: A simple molecule with complex functions. Free Radic Biol Med. 143:176–192. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Wang H, Wu B, Zhang C, Yu H, Li X, Wang Q, Shi X, Fan C, Wang D, et al: Long Noncoding RNA LINC00551 suppresses glycolysis and tumor progression by regulating c-Myc-mediated PKM2 expression in lung adenocarcinoma. Onco Targets Ther. 13:11459–11470. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Y, Zhang D, Zheng T, Yang G, Wang J, Meng F, Liu Y, Zhang G, Zhang L, Han J, et al: lncRNA-SOX2OT promotes hepatocellular carcinoma invasion and metastasis through miR-122-5p-mediated activation of PKM2. Oncogenesis. 9:542020. View Article : Google Scholar : PubMed/NCBI | |
|
Ren J, Li W, Pan G, Huang F, Yang J, Zhang H, Zhou R and Xu N: miR-142-3p modulates cell invasion and migration via PKM2-mediated aerobic glycolysis in colorectal cancer. Anal Cell Pathol (Amst). 2021:99277202021.PubMed/NCBI | |
|
Bian Z, Zhang J, Li M, Feng Y, Wang X, Zhang J, Yao S, Jin G, Du J, Han W, et al: LncRNA-FEZF1-AS1 promotes tumor proliferation and metastasis in colorectal cancer by regulating PKM2 signaling. Clin Cancer Res. 24:4808–4819. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li Q, Pan X, Zhu D, Deng Z, Jiang R and Wang X: Circular RNA MAT2B promotes glycolysis and malignancy of hepatocellular carcinoma through the miR-338-3p/PKM2 axis under hypoxic stress. Hepatology. 70:1298–1316. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gao Y, Yang F, Yang XA, Zhang L, Yu H, Cheng X, Xu S, Pan J, Wang K and Li P: Mitochondrial metabolism is inhibited by the HIF1α-MYC-PGC-1β axis in BRAF V600E thyroid cancer. FEBS J. 286:1420–1436. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou M, He J, Li Y, Jiang L, Ran J, Wang C, Ju C, Du D, Xu X, Wang X, et al: N6-methyladenosine modification of REG1α facilitates colorectal cancer progression via β-catenin/MYC/LDHA axis mediated glycolytic reprogramming. Cell Death Dis. 14:5572023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen M, Cen K, Song Y, Zhang X, Liou YC, Liu P, Huang J, Ruan J, He J, Ye W, et al: NUSAP1-LDHA-Glycolysis-Lactate feedforward loop promotes Warburg effect and metastasis in pancreatic ductal adenocarcinoma. Cancer Lett. 567:2162852023. View Article : Google Scholar : PubMed/NCBI | |
|
Kotowski K, Rosik J, Machaj F, Supplitt S, Wiczew D, Jabłońska K, Wiechec E, Ghavami S and Dzięgiel P: Role of PFKFB3 and PFKFB4 in cancer: Genetic basis, impact on disease development/progression, and potential as therapeutic targets. Cancers (Basel). 13:9092021. View Article : Google Scholar : PubMed/NCBI | |
|
Lai S, Quan Z, Hao Y, Liu J, Wang Z, Dai L, Dai H, He S and Tang B: Long Non-coding RNA LINC01572 promotes hepatocellular carcinoma progression via sponging miR-195-5p to enhance PFKFB4-mediated glycolysis and PI3K/AKT activation. Front Cell Dev Biol. 9:7830882021. View Article : Google Scholar : PubMed/NCBI | |
|
Holman GD: Chemical biology probes of mammalian GLUT structure and function. Biochem J. 475:3511–3534. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wood IS and Trayhurn P: Glucose transporters (GLUT and SGLT): Expanded families of sugar transport proteins. Br J Nutr. 89:3–9. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Yu M, Yongzhi H, Chen S, Luo X, Lin Y, Zhou Y, Jin H, Hou B, Deng Y, Tu L and Jian Z: The prognostic value of GLUT1 in cancers: A systematic review and meta-analysis. Oncotarget. 8:43356–43367. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Macheda ML, Rogers S and Best JD: Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol. 202:654–662. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Shang R, Wang M, Dai B, Du J, Wang J, Liu Z, Qu S, Yang X, Liu J, Xia C, et al: Long noncoding RNA SLC2A1-AS1 regulates aerobic glycolysis and progression in hepatocellular carcinoma via inhibiting the STAT3/FOXM1/GLUT1 pathway. Mol Oncol. 14:1381–1396. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Zhang Q, Song Y, Hao Y, Cui Y, Zhang X, Zhang X, Qin Y, Zhu G, Wang F, et al: Long non-coding RNA SLC2A1-AS1 induced by GLI3 promotes aerobic glycolysis and progression in esophageal squamous cell carcinoma by sponging miR-378a-3p to enhance Glut1 expression. J Exp Clin Cancer Res. 40:2872021. View Article : Google Scholar : PubMed/NCBI | |
|
Cai K, Chen S, Zhu C, Li L, Yu C, He Z and Sun C: FOXD1 facilitates pancreatic cancer cell proliferation, invasion, and metastasis by regulating GLUT1-mediated aerobic glycolysis. Cell Death Dis. 13:7652022. View Article : Google Scholar : PubMed/NCBI | |
|
Song MY, Lee DY, Yun SM and Kim EH: GLUT3 promotes epithelial-mesenchymal transition via TGF-β/JNK/ATF2 signaling pathway in colorectal cancer cells. Biomedicines. 10:18372022. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Y, Zhang Y, Ran F, Liu J, Lin J, Hao X, Ding L and Ye Q: Let-7a-5p inhibits triple-negative breast tumor growth and metastasis through GLUT12-mediated warburg effect. Cancer Lett. 495:53–65. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Brahimi-Horn MC, Chiche J and Pouyssegur J: Hypoxia and cancer. J Mol Med (Berl). 85:1301–1307. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Adams JM, Difazio LT, Rolandelli RH, Luján JJ, Haskó G, Csóka B, Selmeczy Z and Németh ZH: HIF-1: A key mediator in hypoxia. Acta Physiol Hung. 96:19–28. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Nepal M, Choi HJ, Choi BY, Kim SL, Ryu JH, Kim DH, Lee YH and Soh Y: Anti-angiogenic and anti-tumor activity of Bavachinin by targeting hypoxia-inducible factor-1α. Eur J Pharmacol. 691:28–37. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Xu L, Huan L, Guo T, Wu Y, Liu Y, Wang Q, Huang S, Xu Y, Liang L and He X: LncRNA SNHG11 facilitates tumor metastasis by interacting with and stabilizing HIF-1α. Oncogene. 39:7005–7018. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Z, Li G, Li Z, Liu J and Tang Y: TMEM161B-AS1 suppresses proliferation, invasion and glycolysis by targeting miR-23a-3p/HIF1AN signal axis in oesophageal squamous cell carcinoma. J Cell Mol Med. 25:6535–6549. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Q, Zhu Z, Xiao W, Zong G, Wang C, Jiang W, Li K, Shen J, Guo X, Cui J, et al: Hypoxia-induced circRNF13 promotes the progression and glycolysis of pancreatic cancer. Exp Mol Med. 54:1940–1954. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Xia X, Wang S, Ni B, Xing S, Cao H, Zhang Z, Yu F, Zhao E and Zhao G: Hypoxic gastric cancer-derived exosomes promote progression and metastasis via MiR-301a-3p/PHD3/HIF-1α positive feedback loop. Oncogene. 39:6231–6244. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jin L and Zhou Y: Crucial role of the pentose phosphate pathway in malignant tumors. Oncol Lett. 17:4213–4221. 2019.PubMed/NCBI | |
|
Polat IH, Tarrado-Castellarnau M, Bharat R, Perarnau J, Benito A, Cortés R, Sabatier P and Cascante M: Oxidative pentose phosphate pathway enzyme 6-phosphogluconate dehydrogenase plays a key role in breast cancer metabolism. Biology (Basel). 10:852021.PubMed/NCBI | |
|
Lu M, Lu L, Dong Q, Yu G, Chen J, Qin L, Wang L, Zhu W and Jia H: Elevated G6PD expression contributes to migration and invasion of hepatocellular carcinoma cells by inducing epithelial-mesenchymal transition. Acta Biochim Biophys Sin (Shanghai). 50:370–380. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Chen B, Cai T, Huang C, Zang X, Sun L, Guo S, Wang Q, Chen Z, Zhao Y, Han Z, et al: G6PD-NF-κB-HGF signal in gastric cancer-associated mesenchymal stem cells promotes the proliferation and metastasis of gastric cancer cells by upregulating the expression of HK2. Front Oncol. 11:6487062021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen B, Hong Y, Gui R, Zheng H, Tian S, Zhai X, Xie X, Chen Q, Qian Q, Ren X, 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. 13:8042022. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan M, Zhang X, Yue F, Zhang F, Jiang S, Zhou X, Lv J, Zhang Z, Sun Y, Chen Z, et al: CircNOLC1 promotes colorectal cancer liver metastasis by interacting with AZGP1 and sponging miR-212-5p to regulate reprogramming of the oxidative pentose phosphate pathway. Adv Sci (Weinh). 10:e22052292023. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, He Y, Li Y, Li J, Zhao H, Song G, Miyagishi M, Wu S and Kasim V: NeuroD1 promotes tumor cell proliferation and tumorigenesis by directly activating the pentose phosphate pathway in colorectal carcinoma. Oncogene. 40:6736–6747. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tseng CW, Kuo WH, Chan SH, Chan HL, Chang KJ and Wang LH: Transketolase regulates the metabolic switch to control breast cancer cell metastasis via the α-ketoglutarate signaling pathway. Cancer Res. 78:2799–2812. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Qin Z, Xiang C, Zhong F, Liu Y, Dong Q, Li K, Shi W, Ding C, Qin L and He F: Transketolase (TKT) activity and nuclear localization promote hepatocellular carcinoma in a metabolic and a non-metabolic manner. J Exp Clin Cancer Res. 38:1542019. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Zhao X, Yong H, Xu J, Qu P, Qiao S, Hou P, Li Z, Chu S, Zheng J and Bai J: Transketolase promotes colorectal cancer metastasis through regulating AKT phosphorylation. Cell Death Dis. 13:992022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Li Z, Liu L, Wang Q, Li S, Chen D, Hu Z, Yu T, Ding J, Li J, et al: Long noncoding RNA TSLNC8 is a tumor suppressor that inactivates the interleukin-6/STAT3 signaling pathway. Hepatology. 67:171–187. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng X, Guo H, Liu Z, Qin Z, Cong Y, Ren N, Zhang Y and Zhang N: S100A11 activates the pentose phosphate pathway to induce malignant biological behaviour of pancreatic ductal adenocarcinoma. Cell Death Dis. 13:5682022. View Article : Google Scholar : PubMed/NCBI | |
|
Anderson NM, Mucka P, Kern JG and Feng H: The emerging role and targetability of the TCA cycle in cancer metabolism. Protein Cell. 9:216–237. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Woolbright BL, Rajendran G, Harris RA and Taylor JA III: Metabolic flexibility in cancer: Targeting the pyruvate dehydrogenase kinase: Pyruvate dehydrogenase axis. Mol Cancer Ther. 18:1673–1681. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang G, Liu X, Xie J, Meng J and Ni X: PDK-1 mediated Hippo-YAP-IRS2 signaling pathway and involved in the apoptosis of non-small cell lung cancer cells. Biosci Rep. 39:BSR201820992019. View Article : Google Scholar : PubMed/NCBI | |
|
Sun J, Feng M, Zou H, Mao Y and Yu W: Circ_0000284 facilitates the growth, metastasis and glycolysis of intrahepatic cholangiocarcinoma through miR-152-3p-mediated PDK1 expression. Histol histopathol. 38:1129–1143. 2022.PubMed/NCBI | |
|
Wu K, Wang Z, Huang Y, Yao L, Kang N, Ge W, Zhang R and He W: LncRNA PTPRG-AS1 facilitates glycolysis and stemness properties of esophageal squamous cell carcinoma cells through miR-599/PDK1 axis. J Gastroenterol Hepatol. 37:507–517. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
He Z, Li Z, Zhang X, Yin K, Wang W, Xu Z, Li B, Zhang L, Xu J, Sun G, et al: MiR-422a regulates cellular metabolism and malignancy by targeting pyruvate dehydrogenase kinase 2 in gastric cancer. Cell Death Dis. 9:5052018. View Article : Google Scholar : PubMed/NCBI | |
|
Martin-Perez M, Urdiroz-Urricelqui U, Bigas C and Benitah SA: The role of lipids in cancer progression and metastasis. Cell Metab. 34:1675–1699. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bian X, Liu R, Meng Y, Xing D, Xu D and Lu Z: Lipid metabolism and cancer. J Exp Med. 218:e202016062021. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng C, Geng F, Cheng X and Guo D: Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond). 38:272018.PubMed/NCBI | |
|
Carbonetti G, Wilpshaar T, Kroonen J, Studholme K, Converso C, d'Oelsnitz S and Kaczocha M: FABP5 coordinates lipid signaling that promotes prostate cancer metastasis. Sci Rep. 9:189442019. View Article : Google Scholar : PubMed/NCBI | |
|
Yu CW, Liang X, Lipsky S, Karaaslan C, Kozakewich H, Hotamisligil GS, Bischoff J and Cataltepe S: Dual role of fatty acid-binding protein 5 on endothelial cell fate: A potential link between lipid metabolism and angiogenic responses. Angiogenesis. 19:95–106. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ohata T, Yokoo H, Kamiyama T, Fukai M, Aiyama T, Hatanaka Y, Hatanaka K, Wakayama K, Orimo T, Kakisaka T, et al: Fatty acid-binding protein 5 function in hepatocellular carcinoma through induction of epithelial-mesenchymal transition. Cancer Med. 6:1049–1061. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang G, Bonkovsky HL, de Lemos A and Burczynski FJ: Recent insights into the biological functions of liver fatty acid binding protein 1. J Lipid Res. 56:2238–2247. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Lin YX, Wu XB, Zheng CW, Zhang QL, Zhang GQ, Chen K, Zhan Q and An FM: Mechanistic investigation on the regulation of FABP1 by the IL-6/miR-603 signaling in the pathogenesis of hepatocellular carcinoma. Biomed Res Int. 2021:85796582021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Chen Y, Liu Y, Li Q, Luo J, Wang L, Chen Y, Sang C, Zhang W, Ge X, et al: The lncRNA ZFAS1 regulates lipogenesis in colorectal cancer by binding polyadenylate-binding protein 2 to stabilize SREBP1 mRNA. Mol Ther Nucleic Acids. 27:363–374. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu Y, Dong JT, He B, Zou YF, Li XS, Xi CH and Yu Y: LncRNA SNHG16 induces the SREBP2 to promote lipogenesis and enhance the progression of pancreatic cancer. Future Oncol. 15:3831–3844. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Zhang Y, Guan X, Li X, Zhao Z, Gao Y, Zhang X and Chen R: An integrated transcriptomics and proteomics analysis implicates lncRNA MALAT1 in the regulation of lipid metabolism. Mol Cell Proteomics. 20:1001412021. View Article : Google Scholar : PubMed/NCBI | |
|
Corn KC, Windham MA and Rafat M: Lipids in the tumor microenvironment: From cancer progression to treatment. Prog Lipid Res. 80:1010552020. View Article : Google Scholar : PubMed/NCBI | |
|
Che G, Wang W, Wang J, He C, Yin J, Chen Z, He C, Wang X, Yang Y and Liu J: Sulfotransferase SULT2B1 facilitates colon cancer metastasis by promoting SCD1-mediated lipid metabolism. Clin Transl Med. 14:e15872024. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Chen Z, Zhang Y, Yuan P, Liu J, Ding L and Ye Q: MiR-4310 regulates hepatocellular carcinoma growth and metastasis through lipid synthesis. Cancer Lett. 519:161–171. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Zhou Y, Gao H, Wang Y, Cheng Q, Jian S, Ding Q, Gu W, Yao Y, Ma J, et al: LYAR promotes colorectal cancer progression by upregulating FSCN1 expression and fatty acid metabolism. Oxid Med Cell Longev. 2021:99797072021. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Luo J, Mou K, Peng L, Zhou H, Lei Y, Wang H, Zhao Z, Wang J, Wu J, et al: SDPR inhibits TGF-β induced cancer metastasis through fatty acid oxidation regulation in gastric cancer. Int J Biol Sci. 19:2999–3014. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao P, Yuan F, Xu L, Jin Z, Liu Y, Su J, Yuan L, Peng L, Wang C and Zhang G: HKDC1 reprograms lipid metabolism to enhance gastric cancer metastasis and cisplatin resistance via forming a ribonucleoprotein complex. Cancer Lett. 569:2163052023. View Article : Google Scholar : PubMed/NCBI | |
|
Choi YK and Park KG: Targeting glutamine metabolism for cancer treatment. Biomol Ther (Seoul). 26:19–28. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
De Vitto H, Perez-Valencia J and Radosevich JA: Glutamine at focus: Versatile roles in cancer. Tumour Biol. 37:1541–1558. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wu B, Chen Y, Chen Y, Xie X, Liang H, Peng F and Che W: Circ_0001273 downregulation inhibits the growth, migration and glutamine metabolism of esophageal cancer cells via targeting the miR-622/SLC1A5 signaling axis. Thorac Cancer. 13:1795–1805. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Qian CJ, Tong YY, Wang YC, Teng XS and Yao J: Circ_0001093 promotes glutamine metabolism and cancer progression of esophageal squamous cell carcinoma by targeting miR-579-3p/glutaminase axis. J Bioenerg Biomembr. 54:119–134. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Coloff JL, Murphy JP, Braun CR, Harris IS, Shelton LM, Kami K, Gygi SP, Selfors LM and Brugge JS: Differential glutamate metabolism in proliferating and quiescent mammary epithelial cells. Cell metabolism. 23:867–880. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Liu K, Cui J, Xiong J, Wu H, Peng T and Guo Y: Circ-MBOAT2 knockdown represses tumor progression and glutamine catabolism by miR-433-3p/GOT1 axis in pancreatic cancer. J Exp Clin Cancer Res. 40:1242021. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Chen C, Zhao X, Ye H, Zhao Y, Fu Z, Pan W, Zheng S, Wei L, Nong T, et al: HIF-2α regulates non-canonical glutamine metabolism via activation of PI3K/mTORC2 pathway in human pancreatic ductal adenocarcinoma. J Cell Mol Med. 21:2896–2908. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Li B, Xu Y, Qian L, Xu T, Meng G, Li H, Wang Y, Zhang L, Jiang X, et al: GOT2 silencing promotes reprogramming of glutamine metabolism and sensitizes hepatocellular carcinoma to glutaminase inhibitors. Cancer Res. 82:3223–3235. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ghosh P, Vidal C, Dey S and Zhang L: Mitochondria Targeting as an Effective Strategy for Cancer Therapy. Int J Mol Sci. 21:33632020. View Article : Google Scholar : PubMed/NCBI | |
|
Fu H, Zhang J, Chen H, Hou H, Chen H, Xie R, Chen Y, Zhang J, Liu D, Yan L, et al: NPR1 promotes lipid droplet lipolysis to enhance mitochondrial oxidative phosphorylation and fuel gastric cancer metastasis. Adv Sci (Weinh). 37:e032332025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang T, Liang N, Zhang J, Bai Y, Li Y, Zhao Z, Chen L, Yang M, Huang Q, Hu P, et al: OCTN2 enhances PGC-1α-mediated fatty acid oxidation and OXPHOS to support stemness in hepatocellular carcinoma. Metabolism. 147:1556282023. View Article : Google Scholar : PubMed/NCBI | |
|
Mookerjee SA, Gerencser AA, Nicholls DG and Brand MD: Quantifying intracellular rates of glycolytic and oxidative ATP production and consumption using extracellular flux measurements. J Biol Chem. 293:12649–12652. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Lissanu Deribe Y, Sun Y, Terranova C, Khan F, Martinez-Ledesma J, Gay J, Gao G, Mullinax RA, Khor T, Feng N, et al: Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nat Med. 24:1047–1057. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang T, Sun F, Li C, Nan P, Song Y, Wan X, Mo H, Wang J, Zhou Y, Guo Y, et al: MTA1, a novel ATP synthase complex modulator, enhances colon cancer liver metastasis by driving mitochondrial metabolism reprogramming. Adv Sci (Weinh). 10:e23007562023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Tian W, Ge C, Zhang C, Huang Z, Zhang C, Yang Y and Tian H: SNX17 mediates STAT3 activation to promote hepatocellular carcinoma progression via a retromer dependent mechanism. Int J Biol Sci. 21:2762–2779. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang YN, Zeng ZL, Lu J, Wang Y, Liu ZX, He MM, Zhao Q, Wang ZX, Li T, Lu YX, et al: CPT1A-mediated fatty acid oxidation promotes colorectal cancer cell metastasis by inhibiting anoikis. Oncogene. 37:6025–6040. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xu R, Liu Y, Ma L, Sun Y, Liu H, Yang Y, Jin T and Yang D: NQO1/CPT1A promotes the progression of pancreatic adenocarcinoma via fatty acid oxidation. Acta Biochim Biophys Sin (Shanghai). 55:758–768. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun M, Yue Y, Wang X, Feng H, Qin Y, Chen M, Wang Y and Yan S: ALKBH5-mediated upregulation of CPT1A promotes macrophage fatty acid metabolism and M2 macrophage polarization, facilitating malignant progression of colorectal cancer. Exp Cell Res. 437:1139942024. View Article : Google Scholar : PubMed/NCBI | |
|
Hayes JD, Dinkova-Kostova AT and Tew KD: Oxidative stress in cancer. Cancer Cell. 38:167–197. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng Q, Lv C, Qi L, Wang Y, Hao S, Li G, Sun H, Du L, Li J, Wang C, et al: Sodium selenite inhibits cervical cancer progression via ROS-mediated suppression of glucose metabolic reprogramming. Life Sci. 357:1231092024. View Article : Google Scholar : PubMed/NCBI | |
|
Castelli S, De Falco P, Ciccarone F, Desideri E and Ciriolo MR: Lipid Catabolism and ROS in Cancer: A Bidirectional Liaison. Cancers (Basel). 13:54842021. View Article : Google Scholar : PubMed/NCBI | |
|
Tiwari R, Mondal Y, Bharadwaj K, Mahajan M, Mondal S and Sarkar A: Reactive Oxygen Species (ROS) and their profound influence on regulating diverse aspects of cancer: A concise review. Drug Dev Res. 86:e701072025. View Article : Google Scholar : PubMed/NCBI | |
|
Li D, Wang Y, Dong C, Chen T, Dong A, Ren J, Li W, Shu G, Yang J, Shen W, et al: CST1 inhibits ferroptosis and promotes gastric cancer metastasis by regulating GPX4 protein stability via OTUB1. Oncogene. 42:83–98. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Di Gregorio J, Petricca S, Iorio R, Toniato E and Flati V: Mitochondrial and metabolic alterations in cancer cells. Eur J Cell Biol. 101:1512252022. View Article : Google Scholar : PubMed/NCBI | |
|
Vasan K, Werner M and Chandel NS: Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 32:341–352. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pajak B, Siwiak E, Sołtyka M, Priebe A, Zieliński R, Fokt I, Ziemniak M, Jaśkiewicz A, Borowski R, Domoradzki T and Priebe W: 2-Deoxy-d-Glucose and its analogs: From diagnostic to therapeutic agents. Int J Mol Sci. 21:2342019. View Article : Google Scholar : PubMed/NCBI | |
|
Luo W, Guo S, Zhou Y, Zhu J, Zhao J, Wang M, Sang L, Wang B and Chang B: Hepatocellular carcinoma: Novel understandings and therapeutic strategies based on bile acids (Review). Int J Oncol. 61:1172022. View Article : Google Scholar : PubMed/NCBI | |
|
Gagnière J, Raisch J, Veziant J, Barnich N, Bonnet R, Buc E, Bringer MA, Pezet D and Bonnet M: Gut microbiota imbalance and colorectal cancer. World J Gastroenterol. 22:501–518. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Behnam B and Taghizadeh-Hesary F: Mitochondrial metabolism: A new dimension of personalized oncology. Cancers (Basel). 15:40582023. View Article : Google Scholar : PubMed/NCBI |