Open access

Introductory Chapter: Melatonin, the Integrative Molecule within the Human Architecture

Written By

Cristina Manuela Drăgoi and Alina Crenguța Nicolae

Published: 05 November 2018

DOI: 10.5772/intechopen.81071

From the Edited Volume

Melatonin - Molecular Biology, Clinical and Pharmaceutical Approaches

Edited by Cristina Manuela Drăgoi and Alina Crenguţa Nicolae

Chapter metrics overview

1,008 Chapter Downloads

View Full Metrics

1. Introduction

In the healthcare scientific environment, nowadays, researchers are inspired by endogenous springs of molecules that can be reinterpreted, better understood, or completely reconsidered in their function and ability to sustain the human organism in maintaining its homeostasis [1].

Melatonin is such a tremendous molecule acting in the center of the integrative molecular mechanisms of the body, based on interlinkages of the regulatory systems: neural, endocrine, immune, and genetic, all embodying the uniqueness of human architecture [1, 2].

The endogenous indole system represented by biomolecules with indole structure such as tryptophan, serotonin, and, above all, melatonin conducts the integration mechanisms of the organisms in the great informational variety of the environment. Melatonin is responsible for coordinating and synchronizing the expression of the most important physiological effects of the biological rhythm, imposes an order of the biochemical systems functionality and, globally, depicts the molecular logic of living [2, 3].

The indole ring is considered by scientists as a “privileged” biological structure [4, 5], due to its outstanding ability to form organic active compounds with different affinities for endogenous receptors, mainly for G protein-coupled receptors [6]. The indole structure is widely found at all levels of the biological systems as an important component of the biomolecules and natural products, such as the alkaloids from ergot, essential tryptophan amino acid, serotonin, neuromediator, and melatonin, the main hormone secreted by the pineal gland. As a consequence of its biological effects, the indole nucleus is present in the structure of many marketed medicines [7, 8, 9, 10] or dietary supplements [11, 12, 13], as well as in the prototypes of some drugs that are currently under development.

As a constitutive element of proteins, the essential indole amino acid, tryptophan, has the most pronounced hydrophobic character of all the amino acids and forms a specific hydrophobic environment that contributes to the stabilization of the endogenous protein structure, special characteristics regarding membrane fluidity and transmembrane potential [14, 15]. Also, tryptophan is one of the most important indolic endogenous precursors, being involved in the biosynthesis of all endogenous compounds with indole structure: the neurotransmitter serotonin, the pineal hormone melatonin, the neuromodulator and neurotransmitter tryptamine, 5-hydroxytryptophan, and 5-hydroxyindoleacetic acid, as also in the activity of some specific enzymes, cytochrome c peroxidase. Tryptophan depletion is part of the cytotoxic process and antiproliferative cellular mechanism mediated by γ-interferon. Low serum tryptophan concentrations are clinically correlated with the appearance of some pathological infectious, autoimmune, and, not the last, malignant processes [16, 17, 18].

Tryptophan is the precursor of serotonin, a neurotransmitter with indole structure, with vast biological effects. Emergence of imbalances in the serotoninergic metabolism determines the etiology and pathological neuropsychiatric and systemic disorders, including the development of serotonin-secreting tumors [19, 20, 21, 22]. Thus, a more complete overview of tryptophan and serotonin biochemistry and the precise relationships and interactions of these molecules with other endogenous constituents or structures may contribute to the therapeutic understanding and solving many psychiatric, autoimmune, and neoplastic disorders [23, 24, 25].

In particular, melatonin is an indole neurohormone synthesized mainly in the pineal gland, during the night, being also known as the darkness hormone. Melatonin is not exclusively synthesized by the pineal gland; the retina, the skin, and the gastrointestinal tract are only a few other tissues that produce high amounts of melatonin [26].

The direct precursor of melatonin is the serotonin, naturally synthesized in pinealocytes from L-tryptophan. The regulation system of the melatoninergic synthesis is complex, using central and autonomous pathways, so that there are many pathophysiologic situations where the melatonin secretion is deficient. The alteration of the melatoninergic circadian profile [27] is associated with the susceptibility, development, and evolution of a variety of pathologies, the highest incidence of cancer being registered in shift workers, which have a detrimental day-night alternation [28].

On the other hand, small fluctuations in the steady-state levels of the reactive oxygen and nitrogen species concentrations may play a key role in the intracellular signaling, uncontrolled increases of these highly reactive molecules leading to chain reactions mediated by free radicals, which destroy, without discrimination, proteins, lipids, and DNA, resulting, ultimately, in cell death and being the primary or secondary cause of a wide range of diseases [29, 30, 31, 32, 33, 34, 35].

Melatonin was closely analyzed, under all biochemical aspects, considering its antioxidant mechanisms, intrinsic or modulatory at the level of antioxidant enzymes or in connected supplementary scavenging processes, and revealing a unique molecular antioxidant cascade. Its effects were interpreted in conjunction with other endogenous structures or assessed in controlled release formulations, aimed to enhance antioxidant processes and endogenous indole modulatory actions [36, 37, 38].

This molecule was studied under very different circumstances, from interactions with DNA, in association with other therapeutic agents [39], using different animal models [40, 41, 42, 43, 44, 45] and cell lines [46, 47, 48]. The current scientific interest focuses on revealing melatonin actions on major physiological process, as pregnancy or aging [49, 50], determining its modulatory abilities on different stages of fetus evolution, on healthy aging mechanisms, and on preventing neurodegeneration, melatonin receptors being highly expressed at the placenta level, the BBB mainly by P-glycoprotein overexpression, mediating the mother-fetus interchanges and restricting the xenobiotic way to the fragile developing organism [51, 52, 53].

Melatonin also exerts different effects on the glucose metabolism, considering various targets: it stimulates glucose uptake in muscle cells by phosphorylation of insulin receptor substrate-1 through MT2 signaling, MT2 receptors are expressed in hepatocytes, and melatonin therapy elevates glucose release from the liver [54].

The cardiovascular system physiological, pathophysiological, and molecular endogenous mechanisms are highly influenced by diurnal variations, circadian imbalances affecting gene and protein expression, cardiac remodeling, and promoting ischemia/reperfusion damage [55, 56, 57, 58, 59, 60, 61, 62, 63, 64]. Desynchronizations are frequently registered in patients with hypertension, diabetes mellitus, obesity, and metabolic syndrome [65, 66].

Another important research field for melatonin and its derivatives is identifying predictive biomarkers meant to provide extensive control upon pathologic progression and therapy success, markers that are as minimal invasive as possible and readily available [67, 68, 69, 70, 71, 72].

Melatonin is the integrative molecule in the in vivo milieu of every living cell, mediating the integration complex mechanisms of the individual entity into the environment, synchronizes its cyclic processes, and depicts the circadian distribution of physiological and behavioral processes.

References

  1. 1. Duarte CD, Barreiro EJ, Fraga CAM. Privileged structures: A useful concept for the rational design of new lead drug candidates. Mini-Reviews in Medicinal Chemistry. 2007;7:1108-1119
  2. 2. Dragoi CM. Tryptophan, Serotonin, Melatonin—The Spectacular Triad: Physiological, Pathological and Therapeutic Implications of some Bio-Compounds with Indolic Structure. Lap Lambert Academic Publishing; 2013. ISBN-10: 3659404934; ISBN-13: 978-3659404931
  3. 3. Hardeland R. Antioxidative protection by melatonin: Multiplicity of mechanisms from radical detoxification to radical avoidance. Endocrine. 2005;27:119-130
  4. 4. Rad R, Mracec M, Mraceca M, Oprea T. The privileged structures hypothesis for G protein-coupled receptors-some preliminary results. Revue Roumaine de Chimie. 2007;52:853-858
  5. 5. Rodrigues SAF, Barreiro EJ, Fraga CAM. From nature to drug discovery: The indole scaffold as a privileged structure. Mini-Reviews in Medicinal Chemistry. 2009;9:782-793
  6. 6. Pandi-Perumal SR, Srinivasan V, Spence DW, et al. Physiological effects of melatonin: Role of melatonin receptors and signal transduction pathways. Progress in Neurobiology. 2008;85:335-353
  7. 7. Burcea Dragomiroiu GTA, Ginghina O, Radulescu FS, Lupuleasa D, Barca M, Popa DE, et al. In vitro screening of alcohol-induced dose dumping phenomena for controlled release tramadol tablets. Farmácia. 2015;63(5):670-676
  8. 8. Emet M, Ozcan H, Ozel L, et al. A review of melatonin, its receptors and drugs. Eurasian Journal of Medicine. 2016;48:135-141
  9. 9. Burcea Dragomiroiu GTA, Popa DE, Velescu BS, Andrieș A, Ordeanu V, Nicolae AC, et al. Synthesis, characterization and microbiological activity evaluation of novel hard gelatine capsules with cefaclor and piroxicam. Farmácia. 2016;64(6):887-895
  10. 10. Negrei C, Caruntu C, Ginghina O, Burcea Dragomiroiu GTA, et al. Qualitative and quantitative determination of methotrexate polyglutamates in erythrocytes by high performance liquid chromatography. Revista de Chimie (Bucharest). 2015;66(5):607-610
  11. 11. Tincu RC, Cobilinschi C, Tomescu D, Coman L, Tincu I, Diaconu C, et al. Favourable results for L-carnitine use in valproic acid acute poisoning. Farmácia. 2017;65(3):396-400
  12. 12. Arsene AL, Velescu BS, Drăgoi CM, Nicolae AC, Popa DE, Burcea Dragomiroiu GTA, et al. Associated risks correlated to the interaction of conventional oral antidiabetics and dietary supplements. In: Serafinceanu C, Negoiţă O, Elian V, editors. Interdiab, Book Series, 4th International Conference on Interdisciplinary Management of Diabetes Mellitus and its Complications: “Surgical Crossroads with Diabetes Mellitus”. Bucureşti, România: Editura Niculescu; 2018. ISSN: 2393-3488
  13. 13. Gruia V, Aramă C, Mitrea N, Arsene AL, Grădinaru D, Drăgoi CM. The HPLC plasmatic profile of some fat-soluble antioxidant micronutrients (all-trans-retinol, α-tocopherol, coenzyme Q10) in diabetic and dyslipidemic patients. Farmácia. 2009;57(5):630-638
  14. 14. Drăgoi CM, Mitrea N, Arsene AL, Ilie M, Nicolae AC. Jurkat E6.1 cell line studies regarding the effects of some bio-indoles on the membrane fluidity. Farmácia. 2012;60(1):13-20
  15. 15. Drăgoi CM, Mitrea N, Arsene AL, Nicolae AC, Ilie M. In vitro effects of some bio-indoles on the transmembrane potential of Jurkat E6.1 lymphoblasts. Farmácia. 2012;60(2):240-248
  16. 16. Arsene AL, Dragoi CM, Nicolae AC, Popa DE, Burcea Dragomiroiu GTA, Dumitrescu IB, et al. In: Bankovic J, editor. The Challenging Triad: Microbiota, Immune System and Anti-Cancer Drugs “Anti-Cancer Drugs”. Rijeka, Croatia: InTech; 2016. ISBN: 978-953-51-4959-0
  17. 17. Soroceanu V, Rais C, Stefanescu E, Brumărel M, Safta V, Adauji S, et al. Epidemiological and economic aspects of tuberculosis in children. A comparative analysis: Romania vs. the Republic of Moldova. Farmácia. 2016;64(1):152-158
  18. 18. Rais C, Taerel AE, Stefanescu E, Brumărel M, Safta V, Adauji S, et al. Epidemiological aspects of tuberculosis in adults in Romania versus the Republic of Moldova. Farmácia. 2016;64(1):152-158
  19. 19. Sîrbu CA, Drăgoi CM, Nicolae AC, Pleșa CF. History of interferon treatments in multiple sclerosis-60 years of progress. Farmácia. 2017;65(1):14-18
  20. 20. Tarţa-Arsene O, Leanca M, Dică A, Bran E, Rad F, Timnea O, et al. Dietary omega-3 fatty acids supplementation for attention deficit with hyperactivity disorder in epileptic children. Farmácia. 2017;65(4):550-556
  21. 21. Li W, Risacher SL, Huang E, Saykin AJ, et al. Alzheimer’s disease neuroimaging initiative. Type 2 diabetes mellitus is associated with brain atrophy and hypometabolism in the ADNI cohort. Neurology. 2016;87:595-600
  22. 22. Sirbu CA, Furdu-Lungut E, Plesa CF, Nicolae AC, Drăgoi CM. Pharmacological treatment of relapsing remitting multiple sclerosis-where we are? Farmácia. 2016;64(5):651-655
  23. 23. Ginghină O, Burcea Dragomiroiu GTA, Negrei C, Bârcă M. The importance of chemotherapy in the management strategies regarding hepatic metastases in colorectal cancer. Farmácia. 2014;62(3):429-435
  24. 24. Giudice A, Arra C, Turco MC. Review of molecular mechanisms involved in the activation of the Nrf2-ARE signaling pathway by chemopreventive agents. Methods in Molecular Biology. 2010;647:37-74
  25. 25. Ginghină O, Negrei C, Hudiță A, Ioana-Lavric V, Gălățeanu B, Dragomir S, et al. In vitro impact of some natural compounds on HT-29 colorectal adenocarcinoma cells. Farmácia. 2017;65(6):947-953
  26. 26. Reiter RJ, Tan DX, Rosales-Corral S, et al. The universal nature, unequal distribution and antioxidant actions of melatonin and its derivatives. Mini Reviews in Medicinal Chemistry. 2013;13:3730-3734
  27. 27. Peliciari-Garcia RA, Zanquetta MM, Andrade-Silva J, et al. Expression of circadian clock and melatonin receptors within cultured rat cardiomyocytes. Chronobiology International. 2011;28:21-30
  28. 28. Voiculescu SE, Le Duc D, Roșca AE, Zeca V, Chiţimuș DM, Arsene AL, et al. Behavioral and molecular effects of prenatal continuous light exposure in the adult rat. Brain Research. 2016;1650:51-59
  29. 29. Mahal HS, Sharma HS, Mukerjee T. Antioxidant properties of melatonin: A pulse radiolysis study. Free Radical Biology & Medicine. 1999;26:557-565
  30. 30. Tan D-X, Manchester CC, Terron MP, Flores LJ, Reiter RJ. One molecule, many derivatives; a never-ending interaction of melatonin with reactive oxygen and nitrogen species. Journal of Pineal Research. 2007;42:28-42
  31. 31. Tomas-Zapico C, Coto-Montes A. A proposed mechanism to explain the stimulatory effect of melatonin on antioxidative enzymes. Journal of Pineal Research. 2005;39:99-104
  32. 32. Reiter RJ, Mayo JC, Tan D-X, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as anti-oxidant: Under promises but over delivers. Journal of Pineal Research. 2016;61:253-278
  33. 33. Reiter RJ, Rosales-Corral S, Tan DX, et al. Melatonin as a mitochondria-targeted anti-oxidant: One of nature’s best ideas. Cellular and Molecular Life Sciences. 2017;74:3853-3881
  34. 34. Scaiano JC. Exploratory laser flash photolysis study of free radical reactions and magnetic field effects in melatonin chemistry. Journal of Pineal Research. 1995;19:189-195
  35. 35. Tan DX, Reiter RJ, Manchester CC, et al. Chemical and physical properties and potential mechanisms: Melatonin as a broad spectrum anti-oxidant and free radical scavenger. Current Topics in Medicinal Chemistry. 2002;2:181-197
  36. 36. Butu A, Rodino S, Golea D, Butu M, Butnariu M, Negoescu C, et al. Liposomal nanodelivery system for proteasome inhibitor anticancer drug bortezomib. Farmácia. 2015;63(2):224-229
  37. 37. Choi YH, Yu AM. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Current Pharmaceutical Design. 2014;20:793-807
  38. 38. Dinu Pîrvu CE, Aramă CC, et al. Preliminary preformulation studies for a new norfloxacin ruthenium (III) complex with biological activity. Farmácia. 2013;61(2):251-252
  39. 39. Li W, Zhang H, et al. Overcoming ABC transporter-mediated multidrug resistance: Molecular mechanisms and novel therapeutic drug strategies. Drug Resistance Updates. 2016;27:14-29
  40. 40. Arsene AL, Mitrea N, Dragoi CM. Experimental research regarding melatonin’s effects upon the adrenergic innervation of the mouse isolated vas deferens. Farmácia. 2008;56(2):122-127
  41. 41. Nicolae AC, Mitrea N, Drăgoi CM, Constantinescu MZ, Ciofrângeanu C, Bărboi G, et al. Murine studies regarding the variation of oxidative status in serum, hepatic and brain samples, after administration of some CNS active drugs. Farmácia. 2013;61(4):658-669
  42. 42. Arsene AL, Mitrea N, Lupuliasa D, Dragoi CM, Nicolae AC, Dumitrescu IB, et al. Murine models for developing an individualized neuropsychopharmacotherapy based on the behaviour typology. In: Kocabasoglu N, editor. Mental and Behavioural Disorders and Diseases of the Nervous System, “Mood Disorders”. Rijeka, Croatia: InTech; 2013. ISBN: 978-953-51-0959-4
  43. 43. Koksal M, Oguz E, Baba F, et al. Effects of melatonin on testis histology, oxidative stress and spermatogenesis after experimental testis ischemia-reperfusion in rats. European Review for Medical and Pharmacological Sciences. 2012;16:582-588
  44. 44. Arsene AL, Uivarosi V, Mitrea N, Drăgoi CM, Nicolae AC. The binding properties of some novel ruthenium (III) complexes with human serum transferrin. Biopolymers and Cell. 2011;27(2):141-146
  45. 45. Arsene AL, Mitrea N, Cristea A, Drăgoi CM. Experimental research on mice regarding the implication of melatonin in pain management. Farmácia. 2009;57(2):223-228
  46. 46. Nicolae AC, Arsene AL, Vuță V, Popa DE, Sîrbu CA, Burcea Dragomiroiu GTA, et al. In vitro P-gp expression after administration of CNS active drugs. Farmácia. 2016;64(6):844-850
  47. 47. Arsene AL, Uivarosi V, Mitrea N, Drăgoi CM, Nicolae AC. In vitro studies regarding the interactions of some novel ruthenium (III) complexes with double stranded calf thymus deoxyribonucleic acid (DNA). Farmácia. 2016;64(5):712-716
  48. 48. Nicolae AC, Mitrea N, Arsene AL, Constantinescu M, Vuță V, Drăgoi CM. In vitro P-glycoprotein inhibition assay on N2a murine cell line. Farmácia. 2013;61(3):481-491
  49. 49. Hampl R, Bičíková M, Sosvorová L. Hormones and the blood-brain barrier. Hormone Molecular Biology and Clinical Investigation. 2015;21(3):159-164
  50. 50. Nicolae AC, Drăgoi CM, Ceaușu I, Poalelungi C, Iliescu D, Arsene AL. Clinical implications of the indolergic system and oxidative stress in physiological gestational homeostasis. Farmácia. 2015;63(1):46-51
  51. 51. Vollmer C, Weber AP, Wallenfang M, et al. Melatonin pretreatment improves gastric mucosal blood flow and maintains barrier function during hemorrhagic shock in digs. Microcirculation. 2017;24:e12345-e12356. DOI: 10.1111/mice.12345
  52. 52. Tajes M, Ramos-Fernández E, Weng-Jiang X, et al. The blood-brain barrier: Structure, function and therapeutic approaches to cross it. Molecular Membrane Biology. 2014;31:152-167
  53. 53. Alluri H, Wilson RL, Anasooya Shaji C, et al. Melatonin preserves blood-brain barrier integrity via matrix metalloproteinase-9 inhibition. PLoS One. 2016;11:e0154427
  54. 54. Dragoi CM, Arsene AL, Dinu-Pirvu CE, Dumitrescu IB, Popa DE, Burcea Dragomiroiu GTA, et al. Melatonin, a silent regulator of the glucose homeostasis. In: Caliskan M, Kavaklı H, Öz GC, editors. Carbohydrate. Rijeka, Croatia: InTech; 2017. ISBN: 978-953-51-5023-7
  55. 55. He B, Zhao Y, Xu L, et al. The nuclear melatonin receptor ROR alpha is a novel endogenous defender against myocardial ischemia/reperfusion injury. Journal of Pineal Research. 2016;60:313-326
  56. 56. Klishadi MS, Zarei F, Hejazian SH, Moradi A, Hemati M, Safari F. Losartan protects the heart against ischemia reperfusion injury: Sirtuin 3 involvement. Journal of Pharmacy & Pharmaceutical Sciences. 2015;18:112-123
  57. 57. Diaconu CC, Dragoi CM, Bratu OG, et al. New approaches and perspectives for the pharmacological treatment of arterial hypertension. Farmácia. 2018;66(3):408-415
  58. 58. Wiggins-Dohlvik K, Han MS, Stagg HW, et al. Melatonin inhibits thermal injury-induced hyperpermeability in microvascular endothelial cells. Journal of Trauma and Acute Care Surgery. 2014;77:899-905
  59. 59. Khera AV, Kathiresan S. Is coronary atherosclerosis one disease or many? Setting realistic expectations for precision medicine. Circulation. 2017;135:1005-1007
  60. 60. Lochner A, Huisamen B, Nduhirabandi F. Cardioprotective effect of melatonin against ischaemia/reperfusion damage. Frontiers in Bioscience (Elite Edition). 2013;5:305-315
  61. 61. Diaconu CC, Stănescu AMA, et al. Hyperkalemia and cardiovascular diseases: New molecules for the treatment. Revista de Chimie. 2018;69(6):1367-1370
  62. 62. Yang Y, Sun Y, Wei Y, et al. A review of melatonin as a suitable antioxidant against myocardial ischemia-reperfusion injury and clinical heart diseases. Journal of Pineal Research. 2013;57:357-366
  63. 63. Yip HK, Chang YC, Wallace CG, et al. Melatonin treatment improves adipose-derived mesenchymal stem cell therapy for acute lung ischemia-reperfusion injury. Journal of Pineal Research. 2013;54:207-221
  64. 64. Dominguez-Rodriguez A, Abreu-Gonzalez P, Reiter RJ. Melatonin for cardioprotection in ST elevation myocardial infarction: Are we ready for the challenge? Heart. 2017;103:647-648
  65. 65. Drăgoi CM, Nicolae AC, Grigore C, Dinu-Pîrvu CE, Arsene AL. Characteristics of glucose homeostasis and lipidic profile in a hamster metabolic syndrome model, after the co-administration of melatonin and irbesartan in a multiparticulate pharmaceutical formulation. In: Serafinceanu C, Negoiţă O, Elian V, editors. 2nd International Conference on Interdisciplinary Management of Diabetes Mellitus and its Complications—INTERDIAB Proceedings; Editura Niculescu; 2016. pp. 221-229
  66. 66. Mohamad MA, Mitrea N, Nicolae AC, Constantinescu MZ, Drăgoi CM, Arsene AL, et al. The dynamics of adiponectin and leptin on metabolic syndrome patients and age matched healthy subjects. Farmácia. 2014;62(3):532-545
  67. 67. Vlăsceanu AM, Petraru C, Baconi D, Ghica M, Arsene A, Popa L, et al. Quantitative relationships of urinary cotinine levels in smoking diabetic patients. Farmácia. 2015;63(3):349-356
  68. 68. Barbu CG, Arsene AL, Florea S, Albu A, Sirbu A, Martin S, Nicolae AC, Burcea-Dragomiroiu GT, Popa DE, Velescu BS, Dumitrescu IB, Mitrea N, Draganescu D, Lupuliasa D, Spandidos DA, Tsatsakis AM, Dragoi CM, Fica S. Cardiovascular risk assessment in osteoporotic patients using osteoprotegerin as a reliable predictive biochemical marker. Molecular Medicine Reports. 2017;16(5):6059-6067. https://doi.org/10.3892/mmr.2017.7376
  69. 69. Moran C, Beare R, Phan TG, et al. Type 2 diabetes mellitus and biomarkers of neurodegeneration. Neurology. 2015;85:1123-1130
  70. 70. Diaconu C, Bălăceanu A, Moroşan E. Sepsis biomarkers: Past, present and future. Farmácia. 2015;63(6):811-815
  71. 71. Grădinaru D, Mitrea N, Margină D, Arsene AL, Gruia V, Drăgoi CM, et al. Evaluation of serum osteocalcin in elderly patients with type-2 diabetes mellitus. Farmácia. 2009;57(3):331-338
  72. 72. Caivano A, Laurenzana I, et al. High serum levels of extracellular vesicles expressing malignancy-related markers are released in patients with various types of hematological neoplastic disorders. Tumour Biology. 2015;36:9739-9752

Written By

Cristina Manuela Drăgoi and Alina Crenguța Nicolae

Published: 05 November 2018