Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1794 word(s) 1794 2021-09-24 10:53:17 |
2 format correct Meta information modification 1794 2021-09-26 03:30:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Muhammad, J.S. Warburg Effect in Colorectal Carcinogenesis. Encyclopedia. Available online: https://encyclopedia.pub/entry/14548 (accessed on 29 March 2024).
Muhammad JS. Warburg Effect in Colorectal Carcinogenesis. Encyclopedia. Available at: https://encyclopedia.pub/entry/14548. Accessed March 29, 2024.
Muhammad, Jibran Sualeh. "Warburg Effect in Colorectal Carcinogenesis" Encyclopedia, https://encyclopedia.pub/entry/14548 (accessed March 29, 2024).
Muhammad, J.S. (2021, September 24). Warburg Effect in Colorectal Carcinogenesis. In Encyclopedia. https://encyclopedia.pub/entry/14548
Muhammad, Jibran Sualeh. "Warburg Effect in Colorectal Carcinogenesis." Encyclopedia. Web. 24 September, 2021.
Warburg Effect in Colorectal Carcinogenesis
Edit

Colorectal cancer is one of the most leading causes of death worldwide. The Hallmark of colorectal cancer is the increase of glucose uptake and lactate production even in the presence of oxygen, a phenomenon known as the “Warburg effect”. 

colorectal cancer Warburg effect epigenetic alterations ncRNA genetic mutations anti-glycolysis therapy

1. Introduction

Colorectal cancer (CRC) is the fourth most common type of cancer, with more than 1.84 million new cases reported annually. Despite improvements in diagnosis and treatment, CRC causes approximately one million deaths per year and accounts for the third-highest cancer-related deaths worldwide [1]. Recently, anti-cancer drugs targeting dysregulated cancer cell metabolism have been gaining greater attention in the scientific community [2][3]; therefore, understanding the metabolic pathways in CRC cells may provide a key for developing novel diagnostic and therapeutic options to overcome this disease.

Glucose is metabolized by oxidative phosphorylation (OXPHOS) when oxygen is available to normal cells. Under hypoxic conditions, cells undergo anaerobic glycolysis to produce lactate [4], but even in the presence of oxygen, cancer cells adapt to metabolize a high amount of glucose into lactate to fuel uncontrolled cell growth. This phenomenon is the “Warburg effect”, which is a hallmark of nearly all types of cancer, including CRC [5][6]. CRC is a heterogeneous disease in which numerous oncogenes and tumor suppresser genes are mutated in an adenoma-carcinoma sequence that facilitates CRC progression [1]. Several genetic mutations may lead to the upregulation of enzymes and transporters involved in the Warburg effect. Indeed, CRC cells prefer glycolysis over OXPHOS even under normoxic conditions, leading to mitochondrial dysfunction [7]. Still, it is unclear whether OXPHOS is completely turned off by genetic and epigenetic alteration or whether mitochondria continue to function to generate the energy required by the CRC cells. The former might be true because the net ATP yield generated by OXPHOS is higher than that generated by aerobic glycolysis, but glycolysis is 100 times faster [8]. Moreover, glycolysis protects CRC cells against the toxic byproducts of OXPHOS and provides an acidic environment to enhance the cellular uptake of the essential intermediate metabolites required for proper cancer cell growth [9]. Regardless of the significance of the Warburg effect, some types of cancer depend on more than 90% OXPHOS [8], but some cancer metabolisms are a mixture of OXPHOS and glycolysis [9]. Kaldma et al. reported that in situ human CRC cells use glycolysis in the same way as healthy cells; nevertheless, in malignant cells, increased OXPHOS might be due to stimulation of the mitochondrial biogenesis [10].

2. Metabolic Reprogramming in CRC: Genetic Mutations and ncRNA-Mediated Epigenetic Alteration

Recent studies have suggested that numerous genetic mutations and epigenetic alterations causing abnormal activation of several oncogenes (KRAS [11][12], c-Myc [13], PIM1 [14]), and the inactivation of several tumor suppresser genes (APC [15], TP53 [16], SMAD4 [17], PTEN [18]), reprogram the metabolic pathway in CRC, mediating the Warburg effect. For instance, KRAS expression can be downregulated by the overexpression of miR-143, while the lncRNA (glycolysis-associated lncRNA of colorectal cancer) GLCC1 directly increases c-Myc expression [19][20]. In addition, miR-135, miR-150-5p, miR-34a, and miR-21 target APC, TP53, SMAD4, and PTEN expression to promote CRC progression [19][21]. Also, aberrant activation of various signaling pathways, such as the Wnt/β-catenin, FYN-HIF2A, Receptor Tyrosine Kinase (RTK)/Ras GTPase/MAP kinase (MAPK), and PI3K pathways, modulates CRC cell metabolism [22]. Activation of the Wnt/β-catenin pathway accounts for almost 90% of sporadic CRC and is usually associated with a high rate of aerobic glycolysis [15][23]. Also, the Hippo pathway induces glycolysis via upregulation of yes-associated protein 1 [24]. Mutations in transcription factors such as forkhead box (FOX) [6] and HIF1A [5] genes, which could alter the expression of the enzyme-coding genes involved in glycolysis, have been widely reported in CRC. Those enzymes include phosphoglycerate kinase 1 (PGK1) [25], glucose transporter 1 (GLUT1), hexokinase 2 (HK2), pyruvate kinase M2 (PKM2), and lactate dehydrogenase (LDH) [23]. Furthermore, the cytokine-mediated pro-inflammatory microenvironment may enhance the Warburg effect in CRC [26].
Cancer cells have altered epigenetic mechanisms to manipulate the gene expression for abnormal cell growth and metastasis [27]. These epigenetic mechanisms include DNA methylations, histone modifications, and ncRNA-mediated regulation of gene expression [28][29]. Regulation of gene expression by ncRNAs can happen at multiple levels by interacting with DNA, RNA, or proteins. Moreover, the ncRNAs can modulate chromatin structure and the transcription of adjacent or faraway genes. However, the best of the known mechanisms of ncRNA-mediated gene repression is interference with the transcription machinery, which leads to alteration of the recruitment of transcription factors. The ncRNAs mediate transcriptional regulation on an epigenetic level through interaction with chromatin modifiers, either directly via chromatin looping or by sponging a diversity of miRNAs [30]. Recently, accumulating evidence has also proven the role of regulatory ncRNA in metabolic remodeling of CRC [31][32][33]. These molecules can either function as tumor inducers or suppressers by targeting different players of CRC metabolism genes such as transcription factors, enzymes, and transporters of glycolysis. Profiling those ncRNAs in CRC metabolism is further required, paving the way to identify novel targeted therapies and diagnostic, prognostic, and predictive tests.

Mutations cause aberrant expression of c-Myc via different pathways, such as the Wnt and PI3K/AKT/mTOR signaling pathways [34]. When c-Myc is inhibited, various glycolysis enzymes, such as PDK1, LDH, and GLUT1, are downregulated, resulting in tumor growth suppression [13]. The upregulation of c-Myc indirectly promotes glycolysis in CRC cells through the overexpression of polypyrimidine tract binding protein 1 (PTB1)—a protein that plays a role in pre-mRNA splicing. PTB1 facilitates glycolysis by promoting the splicing of PKM2 in CRC cells [35]. In addition, c-Myc can also facilitate CRC progression by upregulating genes relating to other metabolic pathways. For instance, c-Myc has been shown to tip the balance of CRC metabolism from glycolysis toward OXPHOS by upregulating mitochondrial-related proteins such as PGC-1, CPT1A, and TFAM [36]. c-Myc boosts CRC progression through various mechanisms, such as OXPHOS and glycolysis, allowing the identification of novel molecules in its activity. Unfortunately, targeting c-Myc through small molecules such as antibodies is difficult because it is a nuclear protein without a deep surface binding pocket [34]. Therefore, further studies are required to identify other target therapies downstream of c-Myc or examine different epigenetic mechanisms that modulate its expression in CRC cells.

The mRNA splicing of PKM to form PKM1 or PKM2 is mediated by the three heterogeneous nuclear ribonucleoproteins (hnRNPs)—PTB1, hnRNAPA1, and hnRNAPA2. Recently, studies have demonstrated that PKM2 levels can be epigenetically altered by targeting PKM splicers. Both miR-1 and miR-133b have been shown to inhibit CRC progression by targeting PTBP1. When inhibited, PTBP1 tipped the balance of PKM2 towards PKM1, thereby inhibiting glycolysis [37]. In addition, miR-124, miR-137, and miR-340 have been observed to reduce the growth of CRC cells by inhibiting PKM2 splicing, thereby counteracting the Warburg effect [38]. A newly discovered miR-206 has been shown to target hnRNPA1 and inhibit PKM2 splicing. The reduced PKM2 splicing inhibited glycolysis in CRC cells [39]. The lncRNA MEG3 has also been reported as a tumor suppressor in CRC cells by inhibiting the c-Myc expression and indirectly reducing PKM2 activity, resulting in reduced glycolysis activity [40]. Another study showed that PKM2 levels were elevated by the lncRNA FEZF1-AS1, which improves PKM2 stabilization and thus enhances glycolysis in CRC cells [41]. Together, these studies prove that miRNAs and lncRNAs may impair CRC progression by targeting the Warburg effect and altering the PKM2/PKM1 ratio.

With regard to epigenetic regulation, Gregersen et al. reported that HK2 is the main target for miR-143 and that miR-143 loss is significantly associated with increased glycolysis activity in CRC cells [42]. Moreover, lncRNAs (such as MEG3 and KCNQ1OT1) were also reported to regulate HK2 activity [31][40]. Both these lncRNAs play an important role in regulating the Warburg effect in CRC cells. CRC cells overexpressing MEG have shown a reduced expression of HK2 and an inhibition of glycolysis metabolism [40]. On the other hand, KCNQ1OT1 acts as a proteasome inhibitor to increase the stability of HK2, thereby increasing aerobic glycolysis in CRC cells [31].

Inhibition of GLUT1 expression has been shown to be facilitated by a group of anti-cancer therapies, including DT-13, Oridonin, and Oxymatrine [2][3][43], and by butyrate, which functions as a glycolysis inhibitor in CRC cells [44]. However, it is still unclear whether butyrate affects GLUT1 expression as an HDAC inhibitor, thus warranting further studies. These studies, as mentioned earlier, may provide insights into the relationship between GLUT1 inhibition and chemoresistance in CRC cells.

4. Conclusions and Future Research

In summary, CRC cells benefit from the Warburg effect’s ability to enhance their bioenergetic balance and obtain growth-related advantages from glycolysis-derived metabolites. Genetic analyses and ncRNA-mediated epigenetic research have provided insights into the molecular mechanisms of genes involved in regulating the Warburg effect and the development of tumorigenesis. In this review, we have presented molecular insights into the clinical impacts of oncogenic alterations and the effects of overexpression of transcription factors ( KRAS, APC, c-Myc, P53, and HIF1-α), metabolite transporters (GLUT1), and glycolytic enzymes (HK2, PKM2, PDK1, and LDH) on the Warburg effect in CRC cells. For the first time, we have summarized recent pieces of literature showing the importance of miRNAs and lncRNAs as epigenetic mediators regulating the Warburg effect in CRC cells ( Table 1 ). Genetic mutations and epigenetic alterations that deregulate transcription factors, metabolic transporters, and glycolytic enzymes have been associated with poor prognoses and may be associated with chemoradiotherapy resistance in CRC patients. Novel small molecules targeting these enzymes or transporters exert significant anti-proliferative effects. Hence, glycolytic enzymes and metabolite transporters may be used as biomarkers for predicting CRC prognoses and crucial therapeutic targets. Previous studies have demonstrated that the inhibition of epigenetic factors impacts cancer cell metabolism, although further studies are required to fully understand the effectiveness of these inhibitors on the underlying mechanisms in CRC cells. Future studies, particularly translational research, should incorporate ncRNA analysis of epigenomic biomarkers, allowing for personalized treatment using epigenetic modulators. Additionally, combining epigenetic and genetic targeting might be a more effective strategy for delaying CRC progression.

Table 1. Warburg effect-mediating molecules and their associated epigenetic alteration, resistance to chemotherapy, and tested anti-glycolytic drugs for CRC.

Genes Function   Epigenetic Alteration Therapy Resistance Anti-Glycolysis Therapy
Molecule Expression in CRC Effect on Glycolysis
KRAS Oncogenic activator of RAS/MAPK UNC5B-AS1 [45] Upregulated Activating Anti-EGFR [46][47] 3-BrPA [12], ascorbic acid [11]
c-Myc Oncogenic Transcription factor MEG3 [40]
GLCC1 [20]
miR-181d [48]
miR-124 [35]
Downregulated
Upregulated
Upregulated
Downregulated
Inhibitory
Activating
Activating
Inhibitory
N\A vitamin D [40]
Ketamine [49]
Dioscin [50]
HIF1A Hypoxia-inducible transcription factor METTL3 [51]
YTHDF1 [51]
HIFAL [32]
Upregulated
Upregulated
Upregulated
Activating
Activating
Inhibitory
5-FU [52] Rosmarinic acid [53]
APC Tumor suppressor controlling beta-catenin N\A   N\A N\A DT-13 [2]
Metformin [54]
TP53 Transcription factor and tumor suppresser N\A   N\A N\A DCA [16]
FK866 [55]
PKM2 An enzyme of aerobic glycolysis miR-1 [37]
miR-133b [37]
miR-124 [38]
miR-137 [38]
miR-340 [38]
miR-206 [39]
MEG3 [40]
FEZF1-AS1 [41]
miR-122 [56]
Downregulated
Downregulated
Downregulated
Downregulated
Downregulated
Downregulated
Downregulated
Upregulated
Downregulated
Inhibitory
Inhibitory
Inhibitory
Inhibitory
Inhibitory
Inhibitory
Inhibitory
Activating
Inhibitory
Oxaliplatin [56] Butyrate [57]
vitamin C [58]
Oxymatrine [3]
HK2 An enzyme of aerobic glycolysis miR-143 [42]
MEG3 [40]
KCNQ1OT1 [31]
Downregulated
Downregulated
Upregulated
Inhibitory
Inhibitory
Activating
Oxaliplatin [59]
5-FU [59]
N\A
GLUT1 Glucose transporter miR-760 [60]
miR-143 [61]
circDENND4C [60]
METTL3 [62]
Downregulated
Downregulated
Upregulated
Upregulated
Inhibitory
Inhibitory
Activating
Activating
5-FU [44] DT-13 [2]
Oridonin [43]
Oxymatrine [3]
Butyrate [44]

References

  1. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of colorectal cancer: Incidence, mortality, survival, and risk factors. Prz. Gastroenterol. 2019, 14, 89–103.
  2. Wei, X.; Mao, T.; Li, S.; He, J.; Hou, X.; Li, H.; Zhan, M.; Yang, X.; Li, R.; Xiao, J.; et al. DT-13 inhibited the proliferation of colorectal cancer via glycolytic metabolism and AMPK/mTOR signaling pathway. Phytomedicine 2019, 54, 120–131.
  3. Li, X.; Sun, J.; Xu, Q.; Duan, W.; Yang, L.; Wu, X.; Lu, G.; Zhang, L.; Zheng, Y. Oxymatrine Inhibits Colorectal Cancer Metastasis via Attenuating PKM2-Mediated Aerobic Glycolysis. Cancer Manag. Res. 2020, 12, 9503–9513.
  4. Eslami, M.; Sadrifar, S.; Karbalaei, M.; Keikha, M.; Kobyliak, N.M.; Yousefi, B. Importance of the Microbiota Inhibitory Mechanism on the Warburg Effect in Colorectal Cancer Cells. J. Gastrointest. Cancer 2020, 51, 738–747.
  5. Courtnay, R.; Ngo, D.C.; Malik, N.; Ververis, K.; Tortorella, S.M.; Karagiannis, T.C. Cancer metabolism and the Warburg effect: The role of HIF-1 and PI3K. Mol. Biol. Rep. 2015, 42, 841–851.
  6. Dai, W.; Meng, X.; Mo, S.; Xiang, W.; Xu, Y.; Zhang, L.; Wang, R.; Li, Q.; Cai, G. FOXE1 represses cell proliferation and Warburg effect by inhibiting HK2 in colorectal cancer. Cell Commun. Signal. 2020, 18, 7.
  7. Warburg, O.; Wind, F.; Negelein, E. The Metabolism of Tumors in the Body. J. Gen. Physiol. 1927, 8, 519–530.
  8. Chekulayev, V.; Mado, K.; Shevchuk, I.; Koit, A.; Kaldma, A.; Klepinin, A.; Timohhina, N.; Tepp, K.; Kandashvili, M.; Ounpuu, L.; et al. Metabolic remodeling in human colorectal cancer and surrounding tissues: Alterations in regulation of mitochondrial respiration and metabolic fluxes. Biochem. Biophys. Reports 2015, 4, 111–125.
  9. Zheng, J. Energy metabolism of cancer: Glycolysis versus oxidative phosphorylation (Review). Oncol. Lett. 2012, 4, 1151–1157.
  10. Kaldma, A.; Klepinin, A.; Chekulayev, V.; Mado, K.; Shevchuk, I.; Timohhina, N.; Tepp, K.; Kandashvili, M.; Varikmaa, M.; Koit, A.; et al. An in situ study of bioenergetic properties of human colorectal cancer: The regulation of mitochondrial respiration and distribution of flux control among the components of ATP synthasome. Int. J. Biochem. Cell Biol. 2014, 55, 171–186.
  11. El Halabi, I.; Bejjany, R.; Nasr, R.; Mukherji, D.; Temraz, S.; Nassar, F.J.; El Darsa, H.; Shamseddine, A. Ascorbic Acid in Colon Cancer: From the Basic to the Clinical Applications. Int. J. Mol. Sci. 2018, 19, 2752.
  12. Kawada, K.; Toda, K.; Sakai, Y. Targeting metabolic reprogramming in KRAS-driven cancers. Int. J. Clin. Oncol. 2017, 22, 651–659.
  13. Peng, W.; Huang, W.; Ge, X.; Xue, L.; Zhao, W.; Xue, J. Type Iγ phosphatidylinositol phosphate kinase promotes tumor growth by facilitating Warburg effect in colorectal cancer. EBioMedicine 2019, 44, 375–386.
  14. Zhang, M.; Liu, T.; Sun, H.; Weng, W.; Zhang, Q.; Liu, C.; Han, Y.; Sheng, W. Pim1 supports human colorectal cancer growth during glucose deprivation by enhancing the Warburg effect. Cancer Sci. 2018, 109, 1468–1479.
  15. Cha, P.-H.; Hwang, J.-H.; Kwak, D.-K.; Koh, E.; Kim, K.-S.; Choi, K.-Y. APC loss induces Warburg effect via increased PKM2 transcription in colorectal cancer. Br. J. Cancer 2021, 124, 634–644.
  16. Liang, Y.; Hou, L.; Li, L.; Li, L.; Zhu, L.; Wang, Y.; Huang, X.; Hou, Y.; Zhu, D.; Zou, H.; et al. Dichloroacetate restores colorectal cancer chemosensitivity through the p53/miR-149-3p/PDK2-mediated glucose metabolic pathway. Oncogene 2020, 39, 469–485.
  17. Papageorgis, P.; Cheng, K.; Ozturk, S.; Gong, Y.; Lambert, A.W.; Abdolmaleky, H.M.; Zhou, J.-R.; Thiagalingam, S. Smad4 inactivation promotes malignancy and drug resistance of colon cancer. Cancer Res. 2011, 71, 998–1008.
  18. Xiang, S.; Fang, J.; Wang, S.; Deng, B.; Zhu, L. MicroRNA-135b regulates the stability of PTEN and promotes glycolysis by targeting USP13 in human colorectal cancers. Oncol. Rep. 2015, 33, 1342–1348.
  19. Jung, G.; Hernández-Illán, E.; Moreira, L.; Balaguer, F.; Goel, A. Epigenetics of colorectal cancer: Biomarker and therapeutic potential. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 111–130.
  20. Tang, J.; Yan, T.; Bao, Y.; Shen, C.; Yu, C.; Zhu, X.; Tian, X.; Guo, F.; Liang, Q.; Liu, Q.; et al. LncRNA GLCC1 promotes colorectal carcinogenesis and glucose metabolism by stabilizing c-Myc. Nat. Commun. 2019, 10, 3499.
  21. Liu, F.; Di Wang, X. miR-150-5p represses TP53 tumor suppressor gene to promote proliferation of colon adenocarcinoma. Sci. Rep. 2019, 9, 6740.
  22. Fang, S.; Fang, X. Advances in glucose metabolism research in colorectal cancer. Biomed. Rep. 2016, 5, 289–295.
  23. Brown, R.E.; Short, S.P.; Williams, C.S. Colorectal Cancer and Metabolism. Curr. Colorectal Cancer Rep. 2018, 14, 226–241.
  24. Sun, Z.; Zhang, Q.; Yuan, W.; Li, X.; Chen, C.; Guo, Y.; Shao, B.; Dang, Q.; Zhou, Q.; Wang, Q.; et al. MiR-103a-3p promotes tumour glycolysis in colorectal cancer via hippo/YAP1/HIF1A axis. J. Exp. Clin. Cancer Res. 2020, 39, 250.
  25. Nie, H.; Ju, H.; Fan, J.; Shi, X.; Cheng, Y.; Cang, X.; Zheng, Z.; Duan, X.; Yi, W. O-GlcNAcylation of PGK1 coordinates glycolysis and TCA cycle to promote tumor growth. Nat. Commun. 2020, 11, 36.
  26. Liu, Y.; Xiang, F.; Huang, Y.; Shi, L.; Hu, C.; Yang, Y.; Wang, D.; He, N.; Tao, K.; Wu, K.; et al. Interleukin-22 promotes aerobic glycolysis associated with tumor progression via targeting hexokinase-2 in human colon cancer cells. Oncotarget 2017, 8, 25372–25383.
  27. Muhammad, J.S.; Guimei, M.; Jayakumar, M.N.; Shafarin, J.; Janeeh, A.S.; AbuJabal, R.; Eladl, M.A.; Ranade, A.V.; Ali, A.; Hamad, M. Estrogen-induced hypomethylation and overexpression of YAP1 facilitate breast cancer cell growth and survival. Neoplasia 2021, 23, 68–79.
  28. Muhammad, J.S.; Bajbouj, K.; Shafarin, J.; Hamad, M. Estrogen-induced epigenetic silencing of FTH1 and TFRC genes reduces liver cancer cell growth and survival. Epigenetics 2020, 15, 1302–1318.
  29. Tornesello, M.L.; Faraonio, R.; Buonaguro, L.; Annunziata, C.; Starita, N.; Cerasuolo, A.; Pezzuto, F.; Tornesello, A.L.; Buonaguro, F.M. The Role of microRNAs, Long Non-coding RNAs, and Circular RNAs in Cervical Cancer. Front. Oncol. 2020, 10, 150.
  30. Gusic, M.; Prokisch, H. ncRNAs: New Players in Mitochondrial Health and Disease? Front. Genet. 2020, 11, 95.
  31. Chen, C.; Wei, M.; Wang, C.; Sun, D.; Liu, P.; Zhong, X.; Yu, W. Long noncoding RNA KCNQ1OT1 promotes colorectal carcinogenesis by enhancing aerobic glycolysis via hexokinase-2. Aging 2020, 12, 11685–11697.
  32. Zheng, F.; Chen, J.; Zhang, X.; Wang, Z.; Chen, J.; Lin, X.; Huang, H.; Fu, W.; Liang, J.; Wu, W.; et al. The HIF-1α antisense long non-coding RNA drives a positive feedback loop of HIF-1α mediated transactivation and glycolysis. Nat. Commun. 2021, 12, 1341.
  33. Yang, L.; Zhang, Y.; Bao, J.; Feng, J.-F. Long non-coding RNA BCYRN1 exerts an oncogenic role in colorectal cancer by regulating the miR-204-3p/KRAS axis. Cancer Cell Int. 2020, 20, 453.
  34. Satoh, K.; Yachida, S.; Sugimoto, M.; Oshima, M.; Nakagawa, T.; Akamoto, S.; Tabata, S.; Saitoh, K.; Kato, K.; Sato, S.; et al. Global metabolic reprogramming of colorectal cancer occurs at adenoma stage and is induced by MYC. Proc. Natl. Acad. Sci. USA 2017, 114, E7697–E7706.
  35. Taniguchi, K.; Sugito, N.; Kumazaki, M.; Shinohara, H.; Yamada, N.; Matsuhashi, N.; Futamura, M.; Ito, Y.; Otsuki, Y.; Yoshida, K.; et al. Positive feedback of DDX6/c-Myc/PTB1 regulated by miR-124 contributes to maintenance of the Warburg effect in colon cancer cells. Biochim. Biophys. Acta Mol. Basis Dis. 2015, 1852, 1971–1980.
  36. Liu, Q.; Sun, Y.; Fei, Z.; Yang, Z.; Duan, K.; Zi, J.; Cui, Q.; Yu, M.; Xiong, W. Leptin promotes fatty acid oxidation and OXPHOS via the c-Myc/PGC-1 pathway in cancer cells. Acta Biochim. Biophys. Sin. 2019, 51, 707–714.
  37. Taniguchi, K.; Sakai, M.; Sugito, N.; Kumazaki, M.; Shinohara, H.; Yamada, N.; Nakayama, T.; Ueda, H.; Nakagawa, Y.; Ito, Y.; et al. PTBP1-associated microRNA-1 and -133b suppress the Warburg effect in colorectal tumors. Oncotarget 2016, 7, 18940–18952.
  38. Sun, Y.; Zhao, X.; Zhou, Y.; Hu, Y. miR-124, miR-137 and miR-340 regulate colorectal cancer growth via inhibition of the Warburg effect. Oncol. Rep. 2012, 28, 1346–1352.
  39. 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, 465–471.
  40. Zuo, S.; Wu, L.; Wang, Y.; Yuan, X. Long Non-coding RNA MEG3 Activated by Vitamin D Suppresses Glycolysis in Colorectal Cancer via Promoting c-Myc Degradation. Front. Oncol. 2020, 10, 274.
  41. 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. Off. J. Am. Assoc. Cancer Res. 2018, 24, 4808–4819.
  42. Gregersen, L.H.; Jacobsen, A.; Frankel, L.B.; Wen, J.; Krogh, A.; Lund, A.H. MicroRNA-143 down-regulates Hexokinase 2 in colon cancer cells. BMC Cancer 2012, 12, 232.
  43. Yao, Z.; Xie, F.; Li, M.; Liang, Z.; Xu, W.; Yang, J.; Liu, C.; Li, H.; Zhou, H.; Qu, L.-H. Oridonin induces autophagy via inhibition of glucose metabolism in p53-mutated colorectal cancer cells. Cell Death Dis. 2017, 8, e2633.
  44. Geng, H.-W.; Yin, F.-Y.; Zhang, Z.-F.; Gong, X.; Yang, Y. Butyrate Suppresses Glucose Metabolism of Colorectal Cancer Cells via GPR109a-AKT Signaling Pathway and Enhances Chemotherapy. Front. Mol. Biosci. 2021, 8, 634874.
  45. Zhang, Y.; Li, Z.; Lan, Z. Silencing UNC5B antisense lncRNA 1 represses growth and metastasis of human Colon cancer cells via raising miR-622. Artif. Cells Nanomed. Biotechnol. 2020, 48, 60–67.
  46. Ye, H.; Liu, Y.; Wu, K.; Luo, H.; Cui, L. AMPK activation overcomes anti-EGFR antibody resistance induced by KRAS mutation in colorectal cancer. Cell Commun. Signal. 2020, 18, 115.
  47. Bellier, J.; Nokin, M.-J.; Caprasse, M.; Tiamiou, A.; Blomme, A.; Scheijen, J.L.; Koopmansch, B.; MacKay, G.M.; Chiavarina, B.; Costanza, B.; et al. Methylglyoxal Scavengers Resensitize KRAS-Mutated Colorectal Tumors to Cetuximab. Cell Rep. 2020, 30, 1400–1416.
  48. Guo, X.; Zhu, Y.; Hong, X.; Zhang, M.; Qiu, X.; Wang, Z.; Qi, Z.; Hong, X. miR-181d and c-myc-mediated inhibition of CRY2 and FBXL3 reprograms metabolism in colorectal cancer. Cell Death Dis. 2017, 8, e2958.
  49. Hu, J.; Duan, W.; Liu, Y. Ketamine inhibits aerobic glycolysis in colorectal cancer cells by blocking the NMDA receptor-CaMK II-c-Myc pathway. Clin. Exp. Pharmacol. Physiol. 2020, 47, 848–856.
  50. Wu, Z.; Han, X.; Tan, G.; Zhu, Q.; Chen, H.; Xia, Y.; Gong, J.; Wang, Z.; Wang, Y.; Yan, J. Dioscin Inhibited Glycolysis and Induced Cell Apoptosis in Colorectal Cancer via Promoting c-myc Ubiquitination and Subsequent Hexokinase-2 Suppression. Oncol. Targets Ther. 2020, 13, 31–44.
  51. Yang, Z.; Quan, Y.; Chen, Y.; Huang, Y.; Huang, R.; Yu, W.; Wu, D.; Ye, M.; Min, Z.; Yu, B. Knockdown of RNA N6-methyladenosine methyltransferase METTL3 represses Warburg effect in colorectal cancer via regulating HIF-1α. Signal Transduct. Targets Ther. 2021, 6, 89.
  52. Nakagawa, Y.; Kuranaga, Y.; Tahara, T.; Yamashita, H.; Shibata, T.; Nagasaka, M.; Funasaka, K.; Ohmiya, N.; Akao, Y. Induced miR-31 by 5-fluorouracil exposure contributes to the resistance in colorectal tumors. Cancer Sci. 2019, 110, 2540–2548.
  53. Xu, Y.; Han, S.; Lei, K.; Chang, X.; Wang, K.; Li, Z.; Liu, J. Anti-Warburg effect of rosmarinic acid via miR-155 in colorectal carcinoma cells. Eur. J. Cancer Prev. 2016, 25, 481–489.
  54. Cruz-Gil, S.; Sánchez-Martínez, R.; Wagner-Reguero, S.; Stange, D.; Schölch, S.; Pape, K.; Ramírez de Molina, A. A more physiological approach to lipid metabolism alterations in cancer: CRC-like organoids assessment. PLoS ONE 2019, 14, e0219944.
  55. Pan, J.-H.; Zhou, H.; Zhu, S.-B.; Huang, J.-L.; Zhao, X.-X.; Ding, H.; Qin, L.; Pan, Y.-L. Nicotinamide phosphoribosyl transferase regulates cell growth via the Sirt1/P53 signaling pathway and is a prognosis marker in colorectal cancer. J. Cell. Physiol. 2019, 234, 4385–4395.
  56. Wang, X.; Zhang, H.; Yang, H.; Bai, M.; Ning, T.; Deng, T.; Liu, R.; Fan, Q.; Zhu, K.; Li, J.; et al. Exosome-delivered circRNA promotes glycolysis to induce chemoresistance through the miR-122-PKM2 axis in colorectal cancer. Mol. Oncol. 2020, 14, 539–555.
  57. Li, Q.; Cao, L.; Tian, Y.; Zhang, P.; Ding, C.; Lu, W.; Jia, C.; Shao, C.; Liu, W.; Wang, D.; et al. Butyrate Suppresses the Proliferation of Colorectal Cancer Cells via Targeting Pyruvate Kinase M2 and Metabolic Reprogramming. Mol. Cell. Proteom. 2018, 17, 1531–1545.
  58. Aguilera, O.; Muñoz-Sagastibelza, M.; Torrejón, B.; Borrero-Palacios, A.; Del Puerto-Nevado, L.; Martínez-Useros, J.; Rodriguez-Remirez, M.; Zazo, S.; García, E.; Fraga, M.; et al. Vitamin C uncouples the Warburg metabolic switch in KRAS mutant colon cancer. Oncotarget 2016, 7, 47954–47965.
  59. Shi, T.; Ma, Y.; Cao, L.; Zhan, S.; Xu, Y.; Fu, F.; Liu, C.; Zhang, G.; Wang, Z.; Wang, R.; et al. B7-H3 promotes aerobic glycolysis and chemoresistance in colorectal cancer cells by regulating HK2. Cell Death Dis. 2019, 10, 308.
  60. Zhang, Z.-J.; Zhang, Y.-H.; Qin, X.-J.; Wang, Y.-X.; 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, 2387–2400.
  61. Zhao, J.; Chen, Y.; Liu, F.; Yin, M. Overexpression of miRNA-143 Inhibits Colon Cancer Cell Proliferation by Inhibiting Glucose Uptake. Arch. Med. Res. 2018, 49, 497–503.
  62. Chen, H.; Gao, S.; Liu, W.; Wong, C.-C.; Wu, J.; Wu, J.; Liu, D.; Gou, H.; Kang, W.; Zhai, J.; et al. RNA N(6)-Methyladenosine Methyltransferase METTL3 Facilitates Colorectal Cancer by Activating the m(6)A-GLUT1-mTORC1 Axis and Is a Therapeutic Target. Gastroenterology 2021, 160, 1284–1300.
More
Information
Subjects: Biology; Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 427
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 26 Sep 2021
1000/1000