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 + 1374 word(s) 1374 2021-06-25 09:51:16 |
2 format change Meta information modification 1374 2021-07-08 07:58:15 |

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.
Samec, M.; Hassan, S.T. Metabolic Anti-Cancer Effects of Melatonin. Encyclopedia. Available online: https://encyclopedia.pub/entry/11781 (accessed on 28 March 2024).
Samec M, Hassan ST. Metabolic Anti-Cancer Effects of Melatonin. Encyclopedia. Available at: https://encyclopedia.pub/entry/11781. Accessed March 28, 2024.
Samec, Marek, Sherif T.s. Hassan. "Metabolic Anti-Cancer Effects of Melatonin" Encyclopedia, https://encyclopedia.pub/entry/11781 (accessed March 28, 2024).
Samec, M., & Hassan, S.T. (2021, July 07). Metabolic Anti-Cancer Effects of Melatonin. In Encyclopedia. https://encyclopedia.pub/entry/11781
Samec, Marek and Sherif T.s. Hassan. "Metabolic Anti-Cancer Effects of Melatonin." Encyclopedia. Web. 07 July, 2021.
Metabolic Anti-Cancer Effects of Melatonin
Edit

Metabolic reprogramming characterized by alterations in nutrient uptake and critical molecular pathways associated with cancer cell metabolism represents a fundamental process of malignant transformation. Melatonin (N-acetyl-5-methoxytryptamine) is a hormone secreted by the pineal gland. Melatonin primarily regulates circadian rhythms but also exerts anti-inflammatory, anti-depressant, antioxidant and anti-tumor activities. Concerning cancer metabolism, melatonin displays significant anticancer effects via the regulation of key components of aerobic glycolysis, gluconeogenesis, the pentose phosphate pathway (PPP) and lipid metabolism. Melatonin treatment affects glucose transporter (GLUT) expression, glucose-6-phosphate dehydrogenase (G6PDH) activity, lactate production and other metabolic contributors. Moreover, melatonin modulates critical players in cancer development, such as HIF-1 and p53. Taken together, melatonin has notable anti-cancer effects at malignancy initiation, progression and metastasing. Further investigations of melatonin impacts relevant for cancer metabolism are expected to create innovative approaches supportive for the effective prevention and targeted therapy of cancers. 

melatonin cancer metabolism Warburg effect mitochondrial dysfunction anti-inflammatory anti-depressant antioxidant anti-tumor predictive preventive personalized medicine (PPPM/3PM)

1. Introduction

Tumor cell metabolism is characteristically different from that of healthy cells [1]. The ability of cancer cells to modify their metabolism and adapt to nutrient-deprived environments to salvage nutrients and thus build biomass and accelerate proliferation is a well-known feature of malignant transformation [2]. Metabolic reprogramming of cancer cells is a hallmark of tumor development. Metabolic changes in tumor cells are driven by oncogenic mutations, hypoxic conditions, altered molecular signals that upregulate anabolic processes and the inhibition of catabolic cascades [1][3]. Changes in specific pathways, including glycolysis, gluconeogenesis, glutaminolysis, the pentose phosphate pathway (PPP), mitochondrial biogenesis and lipid metabolism, contribute to tumor development, invasion and metastasis [4]. Melatonin (C13H16N2O2; PubChem CID: 896; Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Melatonin; (cited 20 April 2021)), a hormone secreted by the pineal gland, contributes to the regulation of circadian rhythms. Since its discovery (more than 60 years ago), melatonin has been extensively investigated in preclinical and clinical research [5]. Clinically, melatonin is used to manage sleep disorders, jetlag, depressive symptoms and anxiety [6][7]. Importantly, melatonin is a strong antioxidant and can protect organisms from carcinogenesis and neurodegeneration [7].
Moreover, melatonin has oncostatic effects by stimulating apoptosis, regulation of survival signaling, suppression of metastasis and angiogenesis and on the epigenetic machinery contributing to the malignant transformation demonstrated in vitro and in vivo [8][9][10][11]. Significantly, melatonin can attenuate the metabolic reprogramming of cancer cells [12][13]. Indeed, melatonin exerts a wide range of different effects, and its functional chemical groups play a key role in the induced oncostatic properties. This is illustrated by the chemical background, where melatonin belongs to the group of acetamides that is acetamide in which one of the hydrogens joined to the nitrogen atom is substituted by a 2-(5-methoxy-1H-indol-3-yl)ethyl group [14]. Acetamides have previously been reported to exhibit anticancer activities [15]. While experimental research has suggested a broad spectrum of melatonin’s anticancer abilities, the hormone’s impact on cancer metabolism requires further investigation. Understanding the processes behind melatonin’s effects on tumor metabolism can support the introduction of new therapeutic strategies to improve quality of life and prolong the overall survival of cancer patients in the context of preventive, predictive and personalized medicine.

2. Structural and Functional Aspects of Melatonin

Melatonin, or 5-methoxy-N-acetyltryptamine, is a methoxyindole discovered in 1958 that is synthesized in the pineal gland (Figure 1) [16]. Melatonin is synthesized from tryptophan and secreted during the night (dark) phase of the day. Its secretion and synthesis are inhibited during the light phase of the day [7]. Tryptophan, an essential amino acid for melatonin synthesis, is hydroxylated into 5-hydroxytryptophan by tryptophan hydroxylase (TPH). 5-hydroxytryptophan is then converted to serotonin by 5-hydroxytryptophan decarboxylase [17]. Arylalkylamine N-acetyltransferase (AANAT) acetylates serotonin to form acetyl-serotonin and serves as a rate-limiting enzyme that regulates the rhythmic synthesis of melatonin [18][19]. Acetyl-serotonin is then converted to melatonin by acetylserotonin-O-methyltransferase (ASMT). Importantly, AANAT activity depends on cyclic AMP (cAMP) production. Light deficiency leads to norepinephrine release from sympathetic nerve fibers, resulting in cAMP synthesis [20]. Synthesized melatonin is released from the pineal gland into circulation [21]. Even though it is mainly produced in the pineal gland, melatonin is also produced elsewhere. Melatonin production occurs in various other tissues; however, these processes occur independently of circadian rhythms, and the synthesized melatonin is not released into circulation. Therefore, melatonin exists in two pools with different functions [7][22]. Melatonin regulates circadian rhythms, and the suprachiasmatic nucleus (SCN) regulates its circadian release. The SCN receives photic information about the environmental day/night cycle via the retinohypothalamic tract (RHT); melatonin biosynthesis occurs in the absence of light. Concurrently, melatonin controls SCN activity via feedback to its receptors (MT1 and MT2) in the SCN [23][24]. Dysregulation of melatonin-related pathways leads to sleep disorders and various health problems [25]. Moreover, melatonin exerts antioxidant and anti-inflammatory effects [26]. Its antioxidant role is associated with the neutralization of reactive nitrogen (RNS) and oxygen (ROS) species that affect the normal function of cells. Free radical accumulation due to disturbed oxidant-antioxidant machinery results in numerous pathological conditions [27][28]. Recent evidence suggests that melatonin treatment increases superoxide dismutase activity (SOD) and other antioxidant enzymes [29]. Moreover, melatonin stimulates an immune response through its receptors [30]. The immunoregulatory effects of melatonin are mediated by the stimulation of cytokines and acceleration of the T helper immune response. Melatonin promotes the production of interleukins (IL)-1, -6 and -12 by monocytes [31]. Moreover, melatonin supports antigen presentation by macrophages to T cells, resulting in cytotoxic T cell activation and proliferation [32]. Additionally, melatonin contributes to blood pressure regulation and autonomic control of cardiovascular function and has protective roles in various cardiovascular diseases [33]. Several studies reveal that melatonin inhibits carcinogenesis through various mechanisms [8][34][35][36][37]. Melatonin’s anticancer effects include pro-apoptotic [38], antiproliferative [39] and anti-angiogenic activities [10][40][41]. Moreover, melatonin exerts a tumor-suppressive capacity through the modulation of free radical scavenger action and immunoregulation via the activation of anticancer immune cells and the attenuation of T-regulatory cells (Tregs) and cancer-associated fibroblasts (CAF) [35][42].
Figure 1. An overview of melatonin biosynthesis pathway. TPH, tryptophan hydroxylase; 5-HTP-D, 5-hydroxytryptophan decarboxylase; AANAT, arylalkylamine N-acetyltransferase; ASMT, acetylserotonin O-methyltransferase.

3. Expert Recommendations in the Framework of Predictive, Preventive and Personalized (3P) Medicine

Melatonin demonstrates protective effects in sleep disturbances and sleep-related disorders, depression, mitochondrial dysfunction, chronic inflammation as well as their interrelationship highly relevant for carcinogenesis and other related pathologies such as stroke [6][25][43][44][45][46][47][48][49][50]. Based on current experimental research, melatonin is proposed to be used as the pharmacological agents with a multi-functional capacity such as to modulate mitochondrial functions in cancer, among others [51]. Current preclinical evidence suggests that melatonin can modulate different molecular cascades directly connected to the suppression of cancer development and progression. Antitumor efficacy of melatonin mediated by inhibition of metastatic potential was demonstrated in different in vitro model systems [52][53][54]. Even though a number of experimental studies confirmed anticancer abilities of melatonin, massive clinical testing is still absent. The imminent need to identify and investigate novel therapeutic approaches in cancer research determines melatonin as a promising agent targeted on cancer combined with conventional therapies. Further investigation on melatonin’s role in cancer initiation and progression can improve its therapeutical potential in the clinical sphere [55][56][57][58].
The above discussed preclinical research provides valuable insights into the effects of melatonin on tumor metabolism and the Warburg phenotype, an essential step for unrestricted tumor cell proliferation and cancer progression. Melatonin regulates the critical components associated with cancer metabolism, such as GLUTs, LDH, G6PDH, HIF-1 or p53. [59][60][61][62][63][64][65][66][67][68][69][70]. Following notable preclinical achievements in the field, we emphasize the necessity to investigate melatonin effects focused on the clinical needs [71][72][73][74].
To this end, population screening focused on individuals in sub-optimal health conditions prior to the clinical onset of the pathologies followed by cost-effective targeted treatment is considered the optimal approach in the framework of 3P medicine as a concept of medicine of the 21st century [75][76][77].

4. Conclusions

In conclusion, comprehensive knowledge of melatonin’s capacity to regulate tumor metabolism are expected to strongly contribute to the identification of innovative approaches to an improved cancer management. The above-discussed results of preclinical research provide valuable knowledge about the effect of melatonin on tumor metabolism and the Warburg phenotype, an essential step for unrestricted tumor cell proliferation and cancer progression. Despite significant results from preclinical research, we emphasize the need to further investigate the effect of melatonin on cancer processes through the regulation of the Warburg phenotype within the scope of other more complex modalities of cancer research, including clinical investigation.

References

  1. Allison, K.E.; Coomber, B.L.; Bridle, B.W. Metabolic reprogramming in the tumour microenvironment: A hallmark shared by cancer cells and t lymphocytes. Immunology 2017, 152, 175–184.
  2. Sun, X.; Wang, M.; Wang, M.; Yu, X.; Guo, J.; Sun, T.; Li, X.; Yao, L.; Dong, H.; Xu, Y. Metabolic reprogramming in triple-negative breast cancer. Front. Oncol. 2020, 10, 428.
  3. Giannattasio, S.; Mirisola, M.G.; Mazzoni, C. Editorial: Cell stress, metabolic reprogramming, and cancer. Front. Oncol. 2018, 8, 236.
  4. Phan, L.M.; Yeung, S.-C.J.; Lee, M.-H. Cancer metabolic reprogramming: Importance, main features, and potentials for precise targeted anti-cancer therapies. Cancer Biol. Med. 2014, 11, 1–19.
  5. Mayo, J.C.; Cernuda, R.; Quiros, I.; Rodriguez, P.; Garcia, J.I.; Hevia, D.; Sainz, R.M. Understanding the role of melatonin in cancer metabolism. Melatonin Res. 2019, 2, 76–104.
  6. Hansen, M.V.; Andersen, L.T.; Madsen, M.T.; Hageman, I.; Rasmussen, L.S.; Bokmand, S.; Rosenberg, J.; Gögenur, I. Effect of melatonin on depressive symptoms and anxiety in patients undergoing breast cancer surgery: A randomized, double-blind, placebo-controlled trial. Breast Cancer Res. Treat. 2014, 145, 683–695.
  7. Kostoglou-Athanassiou, I. Therapeutic applications of melatonin. Ther. Adv. Endocrinol. Metab. 2013, 4, 13–24.
  8. Li, Y.; Li, S.; Zhou, Y.; Meng, X.; Zhang, J.-J.; Xu, D.-P.; Li, H.-B. Melatonin for the prevention and treatment of cancer. Oncotarget 2017, 8, 39896–39921.
  9. Mociková-Kalická, K.; Bojková, B.; Adámeková, E.; Mníchová-Chamilová, M.; Kubatka, P.; Ahlersová, E.; Ahlers, I. Preventive effect of indomethacin and melatonin on 7,12-dimethybenz/a/anthracene-induced mammary carcinogenesis in female sprague-dawley rats. A preliminary report. Folia Biol. 2001, 47, 75–79.
  10. Orendáš, P.; Kubatka, P.; Bojková, B.; Kassayová, M.; Kajo, K.; Výbohová, D.; Kružliak, P.; Péč, M.; Adamkov, M.; Kapinová, A.; et al. Melatonin potentiates the anti-tumour effect of pravastatin in rat mammary gland carcinoma model. Int. J. Exp. Pathol. 2014, 95, 401–410.
  11. Bojková, B.; Kubatka, P.; Qaradakhi, T.; Zulli, A.; Kajo, K. Melatonin may increase anticancer potential of pleiotropic drugs. Int. J. Mol. Sci. 2018, 19, 3910.
  12. Reiter, R.J.; Sharma, R.; Rosales-Corral, S. Anti-warburg effect of melatonin: A proposed mechanism to explain its inhibition of multiple diseases. Int. J. Mol. Sci. 2021, 22, 764.
  13. Kubatka, P.; Zubor, P.; Busselberg, D.; Kwon, T.K.; Adamek, M.; Petrovic, D.; Opatrilova, R.; Gazdikova, K.; Caprnda, M.; Rodrigo, L.; et al. Melatonin and breast cancer: Evidences from preclinical and human studies. Crit. Rev. Oncol. Hematol. 2018, 122, 133–143.
  14. Zhao, Y.; Ren, J.; Hillier, J.; Jones, M.; Lu, W.; Jones, E.Y. Structural characterization of melatonin as an inhibitor of the wnt deacylase notum. J. Pineal. Res. 2020, 68, e12630.
  15. Rani, P.; Pal, D.; Hegde, R.R.; Hashim, S.R. Acetamides: Chemotherapeutic agents for inflammation-associated cancers. J. Chemother. 2016, 28, 255–265.
  16. Tordjman, S.; Chokron, S.; Delorme, R.; Charrier, A.; Bellissant, E.; Jaafari, N.; Fougerou, C. Melatonin: Pharmacology, functions and therapeutic benefits. Curr. Neuropharmacol. 2017, 15, 434–443.
  17. Zhao, D.; Yu, Y.; Shen, Y.; Liu, Q.; Zhao, Z.; Sharma, R.; Reiter, R.J. Melatonin synthesis and function: Evolutionary history in animals and plants. Front. Endocrinol. 2019, 10, 249.
  18. Saha, S.; Singh, K.M.; Gupta, B.B.P. Melatonin synthesis and clock gene regulation in the pineal organ of teleost fish compared to mammals: Similarities and differences. Gen. Comp. Endocrinol. 2019, 279, 27–34.
  19. Benyassi, A.; Schwartz, C.; Coon, S.L.; Klein, D.C.; Falcón, J. Melatonin synthesis: Arylalkylamine N-acetyltransferases in trout retina and pineal organ are different. Neuroreport 2000, 11, 255–258.
  20. Low, M.J. Neuroendocrinology. In Williams Textbook of Endocrinology, 13th ed.; Melmed, S., Polonsky, K.S., Larsen, P.R., Kronenberg, H.M., Eds.; Elsevier: Philadelphia, PA, USA, 2016; pp. 109–175. ISBN 978-0-323-29738-7.
  21. Cook, J.S.; Sauder, C.L.; Ray, C.A. Melatonin differentially affects vascular blood flow in humans. Am. J. Physiol. Heart Circ. Physiol. 2011, 300, H670–H674.
  22. Reiter, R.J. Circadian and non-circadian melatonin: Influences on glucose metabolism in cancer cells. J. Curr. Sci. Technol. 2020, 10, 85–98.
  23. Hardeland, R. Chronobiology of melatonin beyond the feedback to the suprachiasmatic nucleus—Consequences to melatonin dysfunction. Int. J. Mol. Sci. 2013, 14, 5817–5841.
  24. Dubocovich, M.L. Melatonin receptors: Role on sleep and circadian rhythm regulation. Sleep Med. 2007, 8 (Suppl. S3), 34–42.
  25. Poza, J.J.; Pujol, M.; Ortega-Albás, J.J.; Romero, O. Melatonin in sleep disorders. Neurología 2020.
  26. Ashrafizadeh, M.; Najafi, M.; Kavyiani, N.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Anti-inflammatory activity of melatonin: A focus on the role of NLRP3 inflammasome. Inflammation 2021.
  27. Karaaslan, C.; Suzen, S. Antioxidant properties of melatonin and its potential action in diseases. Curr. Top Med. Chem. 2015, 15, 894–903.
  28. Ahmadi, Z.; Ashrafizadeh, M. Melatonin as a potential modulator of Nrf2. Fundam. Clin. Pharmacol. 2020, 34, 11–19.
  29. Abadi, S.H.M.H.; Shirazi, A.; Alizadeh, A.M.; Changizi, V.; Najafi, M.; Khalighfard, S.; Nosrati, H. The effect of melatonin on superoxide dismutase and glutathione peroxidase activity, and malondialdehyde levels in the targeted and the non-targeted lung and heart tissues after irradiation in xenograft mice colon cancer. Curr. Mol. Pharmacol. 2018, 11, 326–335.
  30. Pohanka, M. Impact of melatonin on immunity: A review. Cent. Eur. J. Med. 2013, 8, 369–376.
  31. Maestroni, G.J.M. Melatonin and the immune system therapeutic potential in cancer, viral diseases, and immunodeficiency states. In The Pineal Gland and Cancer: Neuroimmunoendocrine Mechanisms in Malignancy; Bartsch, C., Bartsch, H., Blask, D.E., Cardinali, D.P., Hrushesky, W.J.M., Mecke, D., Eds.; Springer: Berlin/Heidelberg, Germany, 2001; pp. 384–394. ISBN 978-3-642-59512-7.
  32. Miller, S.C.; Pandi, P.S.R.; Esquifino, A.I.; Cardinali, D.P.; Maestroni, G.J.M. The role of melatonin in immuno-enhancement: Potential application in cancer. Int. J. Exp. Pathol. 2006, 87, 81–87.
  33. Campos, L.A.; Bueno, C.; Barcelos, I.P.; Halpern, B.; Brito, L.C.; Amaral, F.G.; Baltatu, O.C.; Cipolla-Neto, J. Melatonin therapy improves cardiac autonomic modulation in pinealectomized patients. Front. Endocrinol. 2020, 11, 239.
  34. Wang, Y.; Wang, P.; Zheng, X.; Du, X. Therapeutic strategies of melatonin in cancer patients: A systematic review and meta-analysis. OncoTargets Ther. 2018, 11, 7895–7908.
  35. Di Bella, G.; Mascia, F.; Gualano, L.; Di Bella, L. Melatonin anticancer effects: Review. Int. J. Mol. Sci. 2013, 14, 2410–2430.
  36. Wang, T.; Liu, B.; Guan, Y.; Gong, M.; Zhang, W.; Pan, J.; Liu, Y.; Liang, R.; Yuan, Y.; Ye, L. Melatonin inhibits the proliferation of breast cancer cells induced by bisphenol a via targeting estrogen receptor-related pathways. Thorac. Cancer 2018, 9, 368–375.
  37. Hill, S.M.; Frasch, T.; Xiang, S.; Yuan, L.; Duplessis, T.; Mao, L. Molecular mechanisms of melatonin anticancer effects. Integr. Cancer Ther. 2009, 8, 337–346.
  38. Rodriguez, C.; Martín, V.; Herrera, F.; García-Santos, G.; Rodriguez-Blanco, J.; Casado-Zapico, S.; Sánchez-Sánchez, A.M.; Suárez, S.; Puente-Moncada, N.; Anítua, M.J.; et al. Mechanisms involved in the pro-apoptotic effect of melatonin in cancer cells. Int. J. Mol. Sci. 2013, 14, 6597–6613.
  39. Moretti, R.M.; Marelli, M.M.; Maggi, R.; Dondi, D.; Motta, M.; Limonta, P. Antiproliferative action of melatonin on human prostate cancer LNCaP cells. Oncol. Rep. 2000, 7, 347–351.
  40. Gatti, G.; Lucini, V.; Dugnani, S.; Calastretti, A.; Spadoni, G.; Bedini, A.; Rivara, S.; Mor, M.; Canti, G.; Scaglione, F.; et al. Antiproliferative and pro-apoptotic activity of melatonin analogues on melanoma and breast cancer cells. Oncotarget 2017, 8, 68338–68353.
  41. Cheng, J.; Yang, H.-L.; Gu, C.-J.; Liu, Y.-K.; Shao, J.; Zhu, R.; He, Y.-Y.; Zhu, X.-Y.; Li, M.-Q. Melatonin restricts the viability and angiogenesis of vascular endothelial cells by suppressing HIF-1α/ROS/VEGF. Int. J. Mol. Med. 2019, 43, 945–955.
  42. Mortezaee, K.; Potes, Y.; Mirtavoos-Mahyari, H.; Motevaseli, E.; Shabeeb, D.; Musa, A.E.; Najafi, M.; Farhood, B. Boosting immune system against cancer by melatonin: A mechanistic viewpoint. Life Sci. 2019, 238, 116960.
  43. Koklesova, L.; Samec, M.; Liskova, A.; Zhai, K.; Büsselberg, D.; Giordano, F.A.; Kubatka, P.; Golunitschaja, O. Mitochondrial impairments in aetiopathology of multifactorial diseases: Common origin but individual outcomes in context of 3P medicine. EPMA J. 2021, 1–14.
  44. Qian, S.; Golubnitschaja, O.; Zhan, X. Chronic inflammation: Key player and biomarker-set to predict and prevent cancer development and progression based on individualized patient profiles. EPMA J. 2019, 10, 365–381.
  45. Ma, S.; Zhu, L.; Fan, X.; Luo, T.; Liu, D.; Liang, Z.; Hu, X.; Shi, T.; Tan, W.; Wang, Z. Melatonin derivatives combat with inflammation-related cancer by targeting the main culprit STAT3. Eur. J. Med. Chem. 2021, 211, 113027.
  46. Abolhasanpour, N.; Alihosseini, S.; Golipourkhalili, S.; Badalzadeh, R.; Mahmoudi, J.; Hosseini, L. Insight into the effects of melatonin on endoplasmic reticulum, mitochondrial function, and their cross-talk in the stroke. Arch. Med. Res. 2021.
  47. Polivka, J.; Polivka, J.; Pesta, M.; Rohan, V.; Celedova, L.; Mahajani, S.; Topolcan, O.; Golubnitschaja, O. Risks associated with the stroke predisposition at young age: Facts and hypotheses in light of individualized predictive and preventive approach. EPMA J. 2019, 10, 81–99.
  48. Ferracioli-Oda, E.; Qawasmi, A.; Bloch, M.H. Meta-analysis: Melatonin for the treatment of primary sleep disorders. PLoS ONE 2013, 8, e63773.
  49. Abdelgadir, I.S.; Gordon, M.A.; Akobeng, A.K. Melatonin for the management of sleep problems in children with neurodevelopmental disorders: A systematic review and meta-analysis. Arch. Dis. Child. 2018, 103, 1155–1162.
  50. Cho, J.H.; Bhutani, S.; Kim, C.H.; Irwin, M.R. Anti-inflammatory effects of melatonin: A systematic review and meta-analysis of clinical trials. Brain Behav. Immun. 2021, 93, 245–253.
  51. Proietti, S.; Cucina, A.; Minini, M.; Bizzarri, M. Melatonin, mitochondria, and the cancer cell. Cell. Mol. Life Sci. 2017, 74, 4015–4025.
  52. Reiter, R.J.; Rosales-Corral, S.A.; Tan, D.-X.; Acuna-Castroviejo, D.; Qin, L.; Yang, S.-F.; Xu, K. Melatonin, a full service anti-cancer agent: Inhibition of initiation, progression and metastasis. Int. J. Mol. Sci. 2017, 18, 843.
  53. Borin, T.F.; Arbab, A.S.; Gelaleti, G.B.; Ferreira, L.C.; Moschetta, M.G.; Jardim-Perassi, B.V.; Iskander, A.; Varma, N.R.S.; Shankar, A.; Coimbra, V.B.; et al. Melatonin decreases breast cancer metastasis by modulating rho-associated kinase protein-1 expression. J. Pineal Res. 2016, 60, 3–15.
  54. Glenister, R.; McDaniel, K.; Francis, H.; Venter, J.; Jensen, K.; Dusio, G.; Glaser, S.; Meng, F.; Alpini, G. Therapeutic actions of melatonin on gastrointestinal cancer development and progression. Transl. Gastrointest. Cancer 2013, 2, 110–120.
  55. Reiter, R.J.; Sharma, R.; Ma, Q.; Rorsales-Corral, S.; de Almeida Chuffa, L.G. Melatonin inhibits warburg-dependent cancer by redirecting glucose oxidation to the mitochondria: A mechanistic hypothesis. Cell. Mol. Life Sci. 2020, 77, 2527–2542.
  56. Wang, S.-W.; Tai, H.-C.; Tang, C.-H.; Lin, L.-W.; Lin, T.-H.; Chang, A.-C.; Chen, P.-C.; Chen, Y.-H.; Wang, P.-C.; Lai, Y.-W.; et al. Melatonin impedes prostate cancer metastasis by suppressing MMP-13 expression. J. Cell. Physiol. 2021, 236, 3979–3990.
  57. Zharinov, G.M.; Bogomolov, O.A.; Chepurnaya, I.V.; Neklasova, N.Y.; Anisimov, V.N. Melatonin increases overall survival of prostate cancer patients with poor prognosis after combined hormone radiation treatment. Oncotarget 2020, 11, 3723–3729.
  58. Shen, D.; Ju, L.; Zhou, F.; Yu, M.; Ma, H.; Zhang, Y.; Liu, T.; Xiao, Y.; Wang, X.; Qian, K. The inhibitory effect of melatonin on human prostate cancer. Cell. Commun. Signal. 2021, 19, 34.
  59. Blask, D.E.; Dauchy, R.T.; Dauchy, E.M.; Mao, L.; Hill, S.M.; Greene, M.W.; Belancio, V.P.; Sauer, L.A.; Davidson, L. Light exposure at night disrupts host/cancer circadian regulatory dynamics: Impact on the warburg effect, lipid signaling and tumor growth prevention. PLoS ONE 2014, 9, e102776.
  60. Hevia, D.; Gonzalez-Menendez, P.; Fernandez-Fernandez, M.; Cueto, S.; Rodriguez-Gonzalez, P.; Garcia-Alonso, J.I.; Mayo, J.C.; Sainz, R.M. Melatonin decreases glucose metabolism in prostate cancer cells: A 13C stable isotope-resolved metabolomic study. Int. J. Mol. Sci. 2017, 18, 1620.
  61. Sanchez-Sanchez, A.M.; Antolin, I.; Puente-Moncada, N.; Suarez, S.; Gomez-Lobo, M.; Rodriguez, C.; Martin, V. Melatonin cytotoxicity is associated to warburg effect inhibition in ewing sarcoma cells. PLoS ONE 2015, 10, e0135420.
  62. Mi, L.; Kuang, H. Melatonin regulates cisplatin resistance and glucose metabolism through hippo signaling in hepatocellular carcinoma cells. Cancer Manag. Res. 2020, 12, 1863–1874.
  63. He, M.; Zhou, C.; Lu, Y.; Mao, L.; Xi, Y.; Mei, X.; Wang, X.; Zhang, L.; Yu, Z.; Zhou, Z. Melatonin antagonizes nickel-induced aerobic glycolysis by blocking ROS-mediated HIF-1 α/MiR210/ISCU axis activation. Oxid. Med. Cell. Longev. 2020, 2020, 5406284.
  64. Puente-Moncada, N.; Turos-Cabal, M.; Sánchez-Sánchez, A.M.; Antolín, I.; Herrera, F.; Rodriguez-Blanco, J.; Duarte-Olivenza, C.; Rodriguez, C.; Martín, V. Role of glucose metabolism in the differential antileukemic effect of melatonin on wild-type and FLT3-ITD mutant cells. Oncol. Rep. 2020, 44, 293–302.
  65. Hevia, D.; González-Menéndez, P.; Quiros-González, I.; Miar, A.; Rodríguez-García, A.; Tan, D.-X.; Reiter, R.J.; Mayo, J.C.; Sainz, R.M. Melatonin uptake through glucose transporters: A new target for melatonin inhibition of cancer. J. Pineal Res. 2015, 58, 234–250.
  66. Dauchy, R.T.; Hoffman, A.E.; Wren-Dail, M.A.; Hanifin, J.P.; Warfield, B.; Brainard, G.C.; Xiang, S.; Yuan, L.; Hill, S.M.; Belancio, V.P.; et al. Daytime blue light enhances the nighttime circadian melatonin inhibition of human prostate cancer growth. Comp. Med. 2015, 65, 473–485.
  67. Xiang, S.; Dauchy, R.T.; Hauch, A.; Mao, L.; Yuan, L.; Wren, M.A.; Belancio, V.P.; Mondal, D.; Frasch, T.; Blask, D.E.; et al. Doxorubicin resistance in breast cancer is driven by light at night-induced disruption of the circadian melatonin signal. J. Pineal Res. 2015, 59, 60–69.
  68. Dauchy, R.T.; Xiang, S.; Mao, L.; Brimer, S.; Wren, M.A.; Yuan, L.; Anbalagan, M.; Hauch, A.; Frasch, T.; Rowan, B.G.; et al. Circadian and melatonin disruption by exposure to light at night drives intrinsic resistance to tamoxifen therapy in breast cancer. Cancer Res. 2014, 74, 4099–4110.
  69. Chuffa, L.G.A.; Lupi Júnior, L.A.; Seiva, F.R.F.; Martinez, M.; Domeniconi, R.F.; Pinheiro, P.F.F.; Dos Santos, L.D.; Martinez, F.E. Quantitative proteomic profiling reveals that diverse metabolic pathways are influenced by melatonin in an in vivo model of ovarian carcinoma. J. Proteome Res. 2016, 15, 3872–3882.
  70. Mao, L.; Dauchy, R.T.; Blask, D.E.; Dauchy, E.M.; Slakey, L.M.; Brimer, S.; Yuan, L.; Xiang, S.; Hauch, A.; Smith, K.; et al. Melatonin suppression of aerobic glycolysis (Warburg effect), survival signalling and metastasis in human leiomyosarcoma. J. Pineal Res. 2016, 60, 167–177.
  71. Janssens, J.P.; Schuster, K.; Voss, A. Preventive, predictive, and personalized medicine for effective and affordable cancer care. EPMA J. 2018, 9, 113–123.
  72. Golubnitschaja, O. Paradigm change from curative to predictive medicine: Novel strategic trends in Europe. Croat. Med. J. 2009, 50, 596–597.
  73. Liskova, A.; Samec, M.; Koklesova, L.; Giordano, F.A.; Kubatka, P.; Golubnitschaja, O. Liquid biopsy is instrumental for 3PM dimensional solutions in cancer management. J. Clin. Med. 2020, 9, 2749.
  74. Hu, R.; Wang, X.; Zhan, X. Multi-parameter systematic strategies for predictive, preventive and personalised medicine in cancer. EPMA J. 2013, 4, 2.
  75. Crigna, A.T.; Samec, M.; Koklesova, L.; Liskova, A.; Giordano, F.A.; Kubatka, P.; Golubnitschaja, O. Cell-free nucleic acid patterns in disease prediction and monitoring—Hype or hope? EPMA J. 2020, 11, 603–627.
  76. Kunin, A.; Sargheini, N.; Birkenbihl, C.; Moiseeva, N.; Fröhlich, H.; Golubnitschaja, O. Voice perturbations under the stress overload in young individuals: Phenotyping and suboptimal health as predictors for cascading pathologies. EPMA J. 2020, 517–527.
  77. Goncharenko, V.; Bubnov, R.; Polivka, J.; Zubor, P.; Biringer, K.; Bielik, T.; Kuhn, W.; Golubnitschaja, O. Vaginal dryness: Individualised patient profiles, risks and mitigating measures. EPMA J. 2019, 10, 73–79.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 486
Revisions: 2 times (View History)
Update Date: 08 Jul 2021
1000/1000