Biology of radiation-induced mandibular necrosis

0 downloads 0 Views 682KB Size Report
2-3 months after radiotherapy is completed; (b) late effects that occur between 3 .... oxygen species. MLT: melatonin. .... Deng H, Sambrook PJ, Logan RM. The treatment of ... Martin M, Lefaix J, Delanian S. TGF-beta1 and radiation fibrosis: a ...
doi 10.15171/jrb.2018.02

Open Access

Review Article Journal of Radiobiology 2018;5(1):e02

http://www.jradiobiol.com

Biology of radiation-induced mandibular necrosis and the therapeutical perspectives for melatonin Coram Guevara1*, Luis Herrera2, Ricardo Cárdenas3, Freddy Mestre4, Heberto Suarez-Roca3,5 Abstract Radiation therapy is one of the most widely used therapeutic alternatives in the treatment of different types of orofacial cancer. Some anatomical areas in the oral cavity, including jaw bones and salivary glands, are exposed to high doses of radiation, resulting in adverse radiation-induced reactions. One of the most severe radiation-induced reactions is osteoradionecrosis (ORN), which is associated with high morbidity and a significant decrease in the patient’s quality of life. Thus, preventive and therapeutic strategies need to be implemented as to reduce its occurrence. Melatonin, a hormone produced by the pineal gland has anti-tumor, -apoptotic, -oxidant, and anti-inflammatory properties, and has been shown to improve radiation-induced lesions in several organs. Melatonin, when administered preventively, reduces the overproduction of free radicals in peripheral blood cells and apoptosis of normal cells. It also leads to over-expression of pro-inflammatory (DAMPs and TNF) and tolerogenic (TGF-β) mediators that occurs following radiation, thereby modulating the oxidative, necrotic, and fibrotic changes of ORN. Melatonin might be an effective drug for the preventive treatment of ORN. Keywords: Radiotherapy, Ionizing radiation, Osteoradionecrosis, Melatonin

Introduction The effect of x-rays on the human being is the result of interactions that occur at the atomic level (1). These interactions lead to ionization and/or excitation of the orbital electrons, causing the release of energy that, is then deposited in the tissue environment. Some cell groups are more sensitive than others to the ionizing radiation (radiosensitive cells), responding more quickly and at lower doses. For example, reproductive cells are more radiosensitive than nerve cells, and cells with high metabolic rate are more radiosensitive compared to those with lower metabolic rate (2,3). In this sense, ionizing radiation exerts two effects on the tissue; (a) direct effects, in which it acts on the molecules of sensitive cells, and (b) indirect one, where highly reactive free radicals, such as reactive oxygen species (ROS) hydroxyl radical (OH) and hydrogen ions (H), are generated by the radiation (4). These radicals react immediately with neighboring biomolecules. It is estimated that, approximately 60% and 70% of the tissue damage generated by ionizing radiation is caused by OH radicals, which interact with the cell nucleus and the cytoplasm, and as a result, affect the genetic material, organelles, and cellular metabolic processes (5). The main

markers of oxidative damage in cells are the products of lipid peroxidation, products of hydroxylation of DNA, and proteins (6). The deleterious effect of radiation on biological tissues is extremely useful in the treatment of head and neck cancer, the so-called radiotherapy as one of the most used therapeutic alternatives (7). Despite the undoubted clinical utility of radiotherapy in orofacial oncology, and regardless of having the advantage of preserving tissue structure, it causes severe adverse reactions in the oral cavity, being one of the most important cause of osteoradionecrosis (ORN) (8,9). One of the reasons for the high incidence of adverse effects is attributed to the high doses of radiation to which large anatomical areas of the patient are exposed, such as the oral cavity, maxilla, mandible, and salivary glands (10). Radiation-induced inflammation Radiation can cause cell death by various routes, such as necrosis, mitotic catastrophe, apoptosis, autophagy and senescence, and triggering the production of cytokines that can stimulate the activation of various signaling pathways in normal tissue (11). For example, apoptotic cells induce the secretion of tolerogenic cytokines by

Received: 15 January 2018, Accepted: 3 March 2018, Published online: 30 March 2018 Department of Oral Biology, Institute of Research, Faculty of Dentistry, University of Zulia, Venezuela. 2Department of Clinic and Pathology, Institute of Research, Faculty of Dentistry, University of Zulia, Venezuela. 3Section of Neuropharmacology and Neuroscience, Institute of Clinical Research (Dr. Américo Negrette), Faculty of Medicine, University of Zulia, Venezuela. 4Radiotherapy and Nuclear Medicine Service, University Hospital of Maracaibo, Venezuela. 5Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA. *Corresponding Author: Dr. Guevara S. Coram, Department of Oral Biology, Institute of Research, Faculty of Dentistry, University of Zulia, Venezuela. Phone: 00584146305291, Email: [email protected] 1

Guevara et al

Implication for health policy/practice/research/ medical education

Radiation-induced osteoradionecrosis (ORN) in patients undergoing head and neck radiotherapy is considered as one of the most serious complications. It occurs in approximately 20% of patients undergoing local irradiation. In this study, biology of radiation-induced mandibular necrosis and the therapeutical perspectives for melatonin were are reviewed.

macrophages, such as interleukin 10 (IL-10), platelet activating factor, transforming growth factor β (TGF-β), and prostaglandin E2 (PGE2), which exert a suppressive effect of the inflammatory response. On the other hand, the expression of molecular patterns associated with damage (DAMPs) such as the high-mobility group box 1 (HMGB1) and the oxidized DNA product of radiationinduced cell death stimulates the synthesis and secretion of proinflammatory cytokines, such as IL- 1, IL-2, IL-6, tumor necrosis factor (TNF), and interferon gamma (INF-γ) (12). Unlike tolerogenic mediators, all of these mediators have an immunogenic effect and stimulate the inflammatory response. In this sense, the inflammatory response and damage of healthy tissue induced by radiation is carried out by transcription factors such as NF-κβ, protein-kinases (e.g., MAPK), cytokines (IL-1β, IL-4, IL-5, IL-6, IL-10, IL12, IL-18, IL-33, and IFN-γ, TNF-α) and growth factors (TGF-β, bFGF, IGF-1 and PDGF) (13). This response starts with the expression of the DAMPs, which is detected by recognition pattern receptors (PRRs), such as toll-like receptors (TLRs). This, in turn, causes the activation of NF-κβ in various cells, mainly dendritic cells, neutrophils, and macrophages, which activates the signaling pathways for the synthesis of cytokines, and along with TGF-β and IGF-1, stimulates the production of prostaglandins, ROS, and NOS. The upregulation of these factors, after the exposure of high doses of radiation results in a remodeling of the extracellular matrix (ECM) that eventually can affect the tissues and produce pain, atrophy, vascular damage and fibrosis (14). Adverse reactions to radiotherapy depend mainly on the volume and irradiated surface, total dose, fractionation of the treatment (15,16). From the chronological point of view and the fractionation of the treatment, these adverse effects can be divided into two groups: (a) acute effects that are observed during or immediately after treatment, which usually represent an aggravation of pre-existing symptoms. This can be observed from few weeks to up to 2-3 months after radiotherapy is completed; (b) late effects that occur between 3 months to 12 years post-treatment (17,18). The difference between acute and delayed reactions is that, acute reactions appear during treatment and, in general, are reversible while late complications are generally irreversible, leading to permanent disability and decrease in patients’ quality of life (19). 2

Journal of Radiobiology, Vol. 5, No. 1, March 2018

Pathophysiology of osteoradionecrosis Complications that may arise after radiotherapy in patients with head and neck cancer are mainly include decreased quality of masticatory function, dysgeusia, and ORN (20). ORN is considered as the most serious complication (16), occurring in approximately 20% of patients undergoing local irradiation (21). Marx et al (22) has defined ORN as a metabolic and hemostatic deficiency resulting from tissue damage induced by radiation. ORN is characterized by the following sequence: irradiation, hypoxia, hypovascularization, and tissue hypocellularity (23). Irradiation affects the vascular endothelium, causing hyalinization of blood vessels and thrombosis (24). The periosteum becomes fibrous, the osteocytes and osteoclasts suffer necrosis, and subsequent fibrosis of the medullary spaces occurs (23,25). The ability to replace collagen and normal bone cells in irradiated tissues deteriorates extremely, even disappearing completely (23). In the beginning of the 2000s, the theory of radiationinduced fibroatrophy was proposed as the main mechanism of tissue injury in ORN (26). This theory suggests that the main event in the pathophysiology of ORN involves the activation and deregulation of fibroblast activity and the formation of atrophic tissue in previously irradiated areas (Figure 1). An in vivo fibroatrophy study has suggested that TGF-β, through the Smad proteins, plays a fundamental role in the process, since fibrosis is triggered, intensified, and maintained by the participation of the TGF-β1 subunit (27). In vitro studies have also revealed that, this pro-inflammatory cytokine can induce premature expansion and differentiation of a group of pre-fibroblast cells (26-28). During the prefibrotic phase, TGF-β1 secreted by platelets initiates a cascade of events that includes activation and

Figure 1. Pathophysiology of the development of osteoradionecrosis and possible action of melatonin.

Biology of radiation-induced mandibular necrosis

production of macrophages and release of mitogenic factors from fibroblasts (26). During the constitutive and chronic phases, peripheral TGF-β1 produced by myofibroblasts, promotes the initiation and maintenance of the fibrotic process (27). In this setting, overexpression of other inflammatory cytokines and growth factors such as thrombin, IL-1, IL-4, IL-6, TNFα, platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), and connective tissue growth factor (CTGF) triggers the proliferation of smooth muscle tissue and the production of collagen from fibroblasts, leading to the fibrotic process (29). The ORN is usually observed after irradiation at a dose of above 40 Gy (30). These high doses of radiation induce lesions in the vessels due to fibrosis and hyalinization of their wall. The substitution of the normal structure of the vessel wall by fibrosis and unstructured hyaline tissue leads to a functional impairment of the local microcirculation, the vascular lumen narrows and causes ischemic necrosis of the tissue irrigated by the altered vessels (21). On the other hand, in the extracellular space, ROS induces stromal degradation, leukocyte activity (chemotaxis and phagocytosis), and fibroblast activation (31). Within the cell, adaptive responses to oxidative stress are produced through gene activation and protein synthesis, triggering a cascade of events that include DNA repair, cell cycle arrest, and secretion of growth factors, such as TNF -α, plateletderived growth factor (PDGF) and Il-1 (27). However, in vivo studies suggest that the presence of TGF-β1, other pro-inflammatory cytokines, and ROS may be molecular markers of ORN instead of predisposing factors (26,27). In this sense, it has been proposed that the best approach to cope with ORN is the prevention of severe complications associated with radiation therapy in the head and neck by using antioxidants during the peri-irradiation period. One of these antioxidants is melatonin (N-acetyl-5-methoxytryptamine), a hormone produced by the pineal gland in animals. Its main function was thought to be the regulation of sleep and wakefulness, but then it was determined to be a first-line defense molecule against oxidative stress, with remarkable anti-inflammatory properties and possible radioprotective effects (32). Melatonin as a therapeutic alternative for ORN Melatonin is an endogenous compound synthesized by various tissues, including the pineal gland in the human brain (33). Once melatonin is synthesized in the pineal gland, it reaches the cerebrospinal fluid (34-36), and subsequently, since it easily crosses the blood-brain barrier, it quickly accesses the bloodstream (37,38) and other bodily fluids (39), such as bile (40), saliva (41,42), follicular ovarian fluid (43) and semen (44). Melatonin is also synthesized in extra-cellular organs and tissues, including skin, retina and lens, the gastrointestinal tract, cells of the immune and hematopoietic systems, endocrine glands and some reproductive organs (45).

A wide variety of physiological functions have been described for melatonin (46). Melatonin can bind the cytosolic receptor MT3, which is quinone reductase 2 involved in detoxification and protective enzymatic regulation against toxic substances. It modulates immune activity by regulating the expression of antioxidant enzymes by binding orphan nuclear receptors ROR and RZR. Melatonin also exerts effects at the level of the cell surface membrane MT1 and MT2 receptors, which are related with its classical role in the modulation of circadian rhythms, participating in the seasonal variations of reproductive physiology, retinal physiology, regulation of affective and superior cognitive neurobiology, and also other functions as bone growth and regulation of blood pressure. Melatonin is also characterized by antioxidant properties that eliminate free radicals either directly (a property independent of the melatonin binding to its receptors), or indirectly, through stimulating effects on the activities of antioxidant enzymes (46-48), such as superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), glutathione reductase (GR), and catalase (CAT) (49,50). It has been shown that, melatonin directly neutralizes OH radicals, H2O2 and singlet oxygen, and inhibits lipid peroxidation while increasing intracellular levels of glutathione by stimulating the synthesis of the speed-limiting enzyme, γ-glutamylcysteine synthase, which inhibits the nitric oxide and lipoxygenase synthase pro-oxidative enzymes (49). There is evidences that support the stabilizing role of melatonin in microsomal membranes, potentially contributing to resist oxidative damage (51). It has also been observed that melatonin increases the efficiency of the electron transport chain, decreasing the leakage of electrons, thus reducing the generation of free radicals (52). Several studies have demonstrated that, melatonin improves radiation-induced injury in several organs, including spleen (53,54), liver (55,56), lung, colon, spinal cord (57,58), brain (59), femur (60), and salivary glands (61). In this regard, Koc et al (62) reported that melatonin, administered preemptively at a dose of 5 mg/kg, removes free radicals from peripheral blood cells in rats exposed to 5 Gy gamma radiation. It has been shown that, melatonin can have a radioprotective effect preventing cell death by apoptosis. Several in vitro and in vivo studies have provided evidence that melatonin can decrease radiationinduced apoptosis in neurons (63), retina cells (64) and bone marrow cells in rats (65), as well as in mouse thymocytes (66). Similarly, melatonin could modulate the process of apoptosis in the small intestine of mice exposed to gamma radiation at dose of 2.5 Gy (67). Recently, it has been suggested that melatonin plays a role in the reduction of radiation-induced apoptosis in rat cervical spinal cord, by inducing the expression of Bcl-2 family genes (which promote cell survival) and a significant decrease in the expression of the Bax gene (68). Finally, melatonin could Journal of Radiobiology, Vol. 5, No. 1, March 2018

3

Guevara et al

exert additional antiapoptotic effects by blocking the activity of caspase-3 that is necessary for the execution of apoptosis (69). Similarly, melatonin could modulate radiation-induced inflammation (70). It has been shown that melatonin reduces the activation of NF-κβ through the increase of Iκβ (71), as well as the signaling cascade mediated by TLR-4, MyD88, and NF-κβ. The inhibition of NFκβ by melatonin can also reduce the overproduction of leukocytes, adhesion molecules as well as the recruitment of inflammatory cells to the site of injury (72,73). As mentioned above, an immunological response is triggered in the presence of ionizing radiation and it involves the production and release of several mainly proinflammatory cytokines. In this regard, melatonin can exert an antiinflammatory effect by reducing overexpression of proinflammatory cytokines such as TNF-α, IL-1β, and IFN-γ, as well as promoting the Th2-type immune response pattern (74,75). Melatonin can also inhibit the release of TNF-α and IL-8 secreted by neutrophils, which play a leading role in the process of chronic inflammation (76). The administration of melatonin before lung irradiation can reduce the overexpression of TGF-β TNF-α and IL-6 (77). TGF-β has a suppressive effect on the expression of SOD and catalase genes; therefore, a decrease in TGF-β after exposure to ionizing radiation can reduce the effects of oxidative damage (78). As a result, decrease in the expression of the TGF-β gene is an important route for the induction of antioxidant enzymes and the reduction of oxidative damage (79). On the other hand, TGF-β has been shown to play a leading role in the fibrotic process of ORN

(Figure 2), so that melatonin can affect the development of ORN directly by reducing free radicals and oxidative processes or indirectly by inhibiting TGF–β, which will induce the expression of antioxidant enzymes and inhibit ORN-inducing fibrotic process. All of the above evidence supports the therapeutical potential of melatonin as a protector of the adverse injuries induced by radiation. In addition to its antioxidant properties and antiapoptotic effects in normal cells, it has recently been demonstrated that melatonin exerts an antitumor effect, promoting the apoptosis of tumor cells and even preventing tumor angiogenesis (the main mechanism responsible for neoplastic growth and dissemination) (80), stopping the cell cycle, regulating the expression of p53 and Bax upon discharge, and decreasing the expression of Bcl2 (81). Interestingly, in a study of Ehrlich ascitic tumor (EAC) implanted in female mice, it was observed that oral administration of melatonin reduced the viability and volume of neoplastic cells, delayed the progression of the cell cycle, and decreased the DNA content in the cellular interior. Moreover, depressed cell viability indicates that melatonin could be inducing apoptosis in EAC cells (82). Thus, melatonin could be of benefit not only to ameliorate, or even prevent, ORN but also to complement the treatment of the patient’s neoplastic process. Conclusion Radiation-induced ORN represents the most important complication for patients with head and neck cancer who received radiotherapy as the first therapeutic tool. Current knowledge about the pathogenesis of ORN supports the implementation of preventive strategies in the

Figure 2. Inflammation, fibrosis and radiation-induced oxidative damage. Mechanisms of action of melatonin on the effects of radiation on healthy tissue. ROS: reactive oxygen species. MLT: melatonin. PNM: polymorphonuclear. Mφ: macrophages. MEC: extracellular matrix.

4

Journal of Radiobiology, Vol. 5, No. 1, March 2018

Biology of radiation-induced mandibular necrosis

management of this complication. Among these strategies, melatonin emerges as a promising choice for ORN due to its antioxidant properties capable of eliminating the free radicals that are produced in the irradiated tissue as well as its anti-tumor, -apoptotic, -fibrotic, and antiinflammatory effects. Thus, clinical trials should be developed to evaluate the efficacy of melatonin as part of the pharmacological approaches for patients at risk for ORN. Authors’ contribution All the authors contributed equally to the execution and elaboration of the present manuscript. Conflict of interests The authors report no conflicts of interest. The authors alone are responsible for the content and writing of this paper. Ethical considerations Ethical issues (including plagiarism, data fabrication, double publication) have been completely observed by the authors. Also, this article does not contain any studies with human subjects. Funding/Support None declared.

8.

9.

10.

11.

12.

13.

14.

References

1. Coia L, Moyland D. Introduction to Clinical Radiation Oncology. Madison, WI: Medical Physics Publishing; 1998. doi: 10.1259/0007-1285-66-784-387-b 2. Joubert KM, Zimmerman Z, Bencokova J, Gastaldo N, Chavaudra V, Favaudon CF, et al. DNA double-strand break repair defects in syndromes associated with acute radiation response: at least two different assays to predict intrinsic radiosensitivity? Int J Radiat Biol. 2008;84:107125. doi: 10.1080/09553000701797039 3. A. Pavlopoulou, P. Bagos, V. Koutsandrea, A.G. Georgakilas, Molecular determinants of radiosensitivity in normal and tumor tissue: A bioinformatic approach. Cancer Lett. 2017. doi: 10.1016/j.canlet.2017.05.023. 4. Borek C. Antioxidants and Radiation Therapy. American Society for Nutritional Sciences. J Nutr. 2004;134:3207S– 9S. doi: 10.1093/jn/134.11.3207S 5. Devasagayam T, Tilak J, Boloor K, Sane K, Ghaskadbi S, Lele S. Free Radicals and Antioxidants in Human Health: Current Status and Future Prospects. JAPI. 2004;52:794804. doi: 10.1007/978-3-642-30018-9_154 6. Nikitaki V, Nikolov I, Mavragani E, Mladenov A, Mangelis D, Laskaratou G, et al. Measurement of complex DNA damage induction and repair in human cellular systems after exposure to ionizing radiations of varying linear energy transfer (LET). Free Radic Res. 2016;50(sup1):S64-S78. doi:10.1080/10715762.2016.12 32484 7. Thariat J, Hannoun-Levi JM, Sun Myint A, Vuong T, Gerard JP. Past, present, and future of radiotherapy for

15.

16.

17. 18.

19.

20. 21.

the benefit of patients. Nat Rev Clin Oncol. 2013;10:5260. doi:10.1038/nrclinonc.2012.203 Fenwick JD, Pardo-Montero J, Nahum AE, Malik ZI. Impact of schedule duration on head and neck radiotherapy: accelerated tumor repopulation versus compensatory mucosal proliferation. Int J Radiat Oncol Biol Phys. 2012;82(2):1021–30. doi: 10.1016/j. ijrobp.2010.12.048 Cheriex K, Nijhuis T, Mureau M. Osteoradionecrosis of the Jaws: A Review of Conservative and Surgical Treatment Options. J Reconstr Microsurg. 2013; 29:69– 76. doi:10.1055/s-0032-1329923 Vissink A, Jansma J, Spijkervet FKL, Burlage FR, Coppes RP. Oral sequelae of head and neck radiotherapy. Crit Rev Oral Biol Med. 2003;14(3):199–212. doi: 10.1177/154411130301400305 Golden E, Pellicciotta I, Demaria S, Barcellos-Hoff MH, Formenti SC. The convergence of radiation and immunogenic cell death signaling pathways. Front Oncol. 2012;2:88. doi: 10.1177/154411130301400305 Sangiuliano B, Pérez NM, Moreira DF, Belizário JE. Cell death associated molecular-pattern molecules: inflammatory signaling and control. Mediat Inflamm. 2014;2014:821043. doi: 10.1155/2014/821043 Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, et al. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm. 2015;12(1):14. doi: 10.1186/s12950-015-0058-3 Bentzen SM. Preventing or reducing late side effects of radiation therapy: radiobiology meets molecular pathology. Nat Rev Cancer. 2006;6(9):702–713. doi: 10.1038/nrc1950 Dörr W, Hamilton CS, Boyd T, Reed B, Denham JW. Radiation induced changes in cellularity and proliferation in human oral mucosa. Int J Radiat Oncol Biol Phys.  2002;52(4):911–917. doi: 10.1016/S03603016(01)02721-3 Tolentino Ede S, Centurion BS, Ferreira LH, Souza AP, Damante JH, Rubira-Bullen IR. Oral adverse effects of head and neck radiotherapy: Literature review and suggestion of a clinical oral care guideline for irradiated patients. J Appl Oral Sci.2011;19:448–54. doi: 10.1590/ S1678-77572011000500003 Otmani N. Oral and maxillofacial side effects of radiation therapy on children.  J Can Dent Assoc.2007;73(3):257–261. Albuquerque DF, de Souza Tolentino E, Amado FM, Arakawa C, Chinellato LE. Evaluation of halitosis and sialometry in patients submitted to head and neckradiotherapy. Med Oral Patol Oral Cir Bucal. 2010;15:e850–4. doi: 10.4317/medoral.15.e850 Escoda-Francolı J, Rodríguez-Rodríguez A, PérezGarcía S, Gargallo-Albiol J, Gay-Escoda C. Dental implications in oral cáncer patients. Med Oral Patol Oral Cir Bucal. 2011;16:e508–13. doi: 10.4317/medoral.16. e508 Deng H, Sambrook PJ, Logan RM. The treatment of oral cancer: An overview for dental professionals. Aust Dent J. 2011;56:244–52. doi: 10.1111/j.1834-7819.2011.01349.x Blery P, Espitalier F, Hays A, Crauste E, Demarquay C, Pilet

Journal of Radiobiology, Vol. 5, No. 1, March 2018

5

Guevara et al

22.

23.

24.

25.

26.

27.

28.

29. 30.

31. 32.

33.

34.

35.

6

P, et al. Development of mandibular osteoradionecrosis in rats: Importance of dental extraction. Journal of Cranio-Maxillo-Facial Surgery. 2015; 43: 1829e1836. doi: 10.1016/j.jcms.2015.08.016 Marx RE, Johnson RP. Studies in the radiobiology of osteoradionecrosis and their clinical significance. Oral Surg Oral Med Oral Pathol. 1987;64(4):379–90. doi: 10.1016/0030-4220(87)90136-8 De Felice F, Musio D, Tombolini V, Osteoradionecrosis and intensity modulated radiation therapy: An overview. Critical Reviews in Oncology/Hematology. 2016;107:39–43. doi: 10.1016/j.critrevonc.2016.08.017 Barasch A, Epstein JB, Foong WC, Clayman L. Intralesional chemotherapy for head and neck carcinoma: A review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2004;97:307-11. doi: 10.1016/j.tripleo.2003.12.016 Chandra A, Lin T, Tribble MB, Zhu J, Altman AR, Tseng W-J. PTH1e34 alleviates radiotherapy-induced local bone loss by improving osteoblast and osteocyte survival. Bone. 2014;67:33e40. doi: 10.1016/j.bone.2014.06.030 Delanian S, Lefaix JL. The radiation-induced fibroatrophic process: therapeutic perspective via the antioxidant pathway. Radiother Oncol. 2004;73:119– 131. doi: 10.1016/j.radonc.2004.08.021. Martin M, Lefaix J, Delanian S. TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys. 2000;47:277–290. doi: 10.1016/S0360-3016(00)00435-1 Lyons A, Ghazali N. Osteoradionecrosis of the jaws: current understanding of its pathophysiology and treatment. Br J Oral Maxillofac Surg. 2008;46:653–660. doi: 10.1016/j.bjoms.2008.04.006 Lefaix J-L, Daburon F. Diagnosis of acute localized irradiation lesions: a review of the French experimental experience. Health Phys. 1998;75:375–84. Stére G., Boysen M. Mandibular osteoradionecrosis: clinical behaviour and diagnostic aspects. Clin. Otolaryngol. 2000;25:378-384. doi: 10.1046/j.13652273.2000.00367.x Denham J, Hauer-Jensen M. The radiotherapeutic injury—a complex wound. Radiother Oncol. 2002;63:129–45. doi: 10.1016/S0167-8140(02)00060-9 Najafi A, Shirazi E, Motevaseli G, Geraily F, Norouzi M, Heidari S, et al. The melatonin immunomodulatory actions in radiotherapy. Biophys Rev. 2017; 9:139–148. doi: 10.1007/s12551-017-0256-8 Vijayalaxmi V, Reiter R, Tan D, Herman T, Thomas C, Melatonin as a radioprotective agent: a review. International Journal of Radiation Oncology Biology Physics. 2004; 59(3):639–653. doi: 10.1016/j. ijrobp.2004.02.006 Rousseau A, Petren S, Plannthin J, Eklundh T, Nordin C. Serum and cerebrospinal fluid concentrations of melatonin: a pilot study in healthy male volunteers. Journal of Neural Transmission. 1999;106(9-10):883– 888. doi: 10.1007/s007020050208 Leston J, Harthe C, Brun J. Melatonin is released in the third ventricle in humans. A study in movement

Journal of Radiobiology, Vol. 5, No. 1, March 2018

36.

37.

38. 39.

40.

41. 42.

43.

44.

45. 46. 47.

48. 49.

50.

disorders. Neurosci Lett. 2010;469(3):294-97. doi: 10.1016/j.neulet.2009.12.008 Hardeland R, Cardinali D, Srinivasan V, Spence D, Brown G, Pandi-Perumal S. Melatonin-A pleiotropic, orchestrating regulator molecule. Prog Neurobiol. 2011;93(3): 350–384. doi: 10.1016/j. pneurobio.2010.12.004 Pardridge W, Mietus L. Transport of albumin bound melatonin through the blood-brain barrier. J Neurochem. 1980;34(6):1761-3. doi: 10.1111/j.14714159.1980.tb11272.x KennawayD, Voultsios A. Circadian rhythm of free melatonin in human plasma. J Clin Endocrinol Metab. 1998;83(3):1013-5. doi: 10.1210/jcem.83.3.4636 Reiter R. Normal patterns of melatonin levels in the pineal gland and body fluids of humans and experimental animals. J Neural Transm (Vienna). 1986;21;35– 54. doi: 10.1007/978-3-642-59512-7_2 Tan D, Manchester L, Reiter R, Qi W, Hanes M, Farley N. High physiological levels of melatonin in the bile of mammals. Life Sci. 1999;65(23):2523–2529. doi: 10.1016/S0024-3205(99)00519-6 Vakkuri O. Diurnal rhythm of melatonin in human saliva. Acta Physiologica Scandinavica 1985;124(3):40912. doi: 10.1111/j.1748-1716.1985.tb07676.x Nowak R, McMillen I, Redman J, Short R. The correlation between serum and salivary melatonin concentrations and urinary 6-hydroxymelatonin sulphate excretion rates: two non-invasive techniques for monitoring human circadian rhythmicity. Clinical Endocrinology. 1987;27(4):445-52. doi: 10.1111/j.13652265.1987.tb01172.x Brzezinski A, Seibel M, Lynch H. Melatonin in human preovulatory follicular fluid. Journal of Clinical Endocrinology and Metabolism. 1987;64(4):865-7. doi: 10.1210/jcem-64-4-865 Bornman M, Oosthuizen J, Barnard H, Schulenburg G, Boomker D, Reif S. Melatonin and sperm motility. Andrologia. 1989;21(5):483-5. doi: 10.1111/j.14390272.1989.tb02456.x Luchetti F, Canonico B, BettiM. Melatonin signaling and cell protection function. FASEB J 2010; 24(10): 3603– 3624. doi: 10.1096/fj.10-154450 Reiter RJ, Tan DX, Galano A: Melatonin: Exceeding expectations. Physiology (Bethesda) 2014; 29: 325‑333. doi: 10.1152/physiol.00011.2014 Shirazi A, Ghobadi G, Ghazi-Khansari M. A radiobiological review on melatonin: a novel radioprotector. J Radiat Res. 2007;48(4):263-72. doi: 10.1269/jrr.06070 Rodriguez C, Mayo J, Sainz R. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res. 2004; 36(1): 1–9. doi: 10.1046/j.1600-079X.2003.00092.x Karbownik M, Reiter R. Antioxidative effects of melatoninin protection against cellular damage caused by ionizing radiation (44547). Exp Biol Med. 2000; 225(1): 9–22. doi: 10.1111/j.1525-1373.2000.22502.x Reiter R. Oxidative damage in the central nervous system: protection by melatonin. Prog Neurobiol. 1998;

Biology of radiation-induced mandibular necrosis

56(3): 359–384. doi: 10.1016/S0301-0082(98)00052-5 51. Karbownik M, Garcia J, Lewinski A, Reiter R. Carcinogen-induced, free radical-mediated reduction in microsomal membrane fluidity: reversal by indole3-propionic acid. J Bioenerg Biomembr. 2001; 33(1) 73–78. doi: 10.1023/A:1005628808688 52. Reiter R, Tan D, Manchester L, Qi W. Biochemical reactivity of melatonin with reactive oxygen and nitrogen species: a review of the evidence. Cell Biochem Biophys. 2001; 34(2): 237–256. doi: 10.1385/CBB:34:2:237 53. Sharma S, Haldar C. Melatonin prevents X-ray irradiation induced oxidative damage in peripheral blood and spleen of the seasonally breeding rodent, Funambulus pennanti during reproductively active phase. Int J Radiat Biol. 2006; 82(6): 411–419. doi: 10.1080/09553000600774105 54. Sharma S, Haldar C, Chaube S. Effect of exogenous melatonin on X-ray induced cellular toxicity in lymphatic tissue of Indian tropical male squirrel, Funambulus pennant. Int J Radiat Biol. 2008;84(5):363–374. doi: 10.1080/09553000802029894 55. Taysi S, Koc M, Buyukokuroglu M, Altinkaynak K, Sahin Y. Melatonin reduces lipid peroxidation and nitricoxide during irradiation-induced oxidative injury in the rat liver. J Pineal Res. 2003;34(3):173-7. doi: 10.1034/j.1600079X.2003.00024.x 56. Shirazi A, Mihandoost E, Ghobadi G, Mohseni M, Ghazi-Khansari M. Evaluation of radio-protective effect of melatonin on whole body irradiation induced liver tissue damage. Cell J. 2013;14(4):292-7. doi: 10.1016/j. ejmp.2011.11.007 57. Aghazadeh S, Azarnia M, Shirazi A, Mahdavi S, Zangii B. Melatonin as a protective agent in spinal cord damage after gamma irradiation. Rep Pract Oncol Radiother. 2007;12(2):95-9. 58. Haddadi G, Shirazi A, Sepehrizadeh Z, Mahdavi S, Haddadi M. Radioprotective effect of melatonin on the cervical spinal cord in irradiated rats. Cell J. 2013; 14(4): 246-53. doi: 10.1504/IJLR.2010.037665 59. Erol F, Topsakal C, Ozveren M. Protective effects of melatonin and vitamin E in brain damage due to gamma radiation: an experimental study. Neurosurg Rev. 2004; 27(1): 65–69. doi: 10.1007/s10143-003-0291-8 60. Çakir ZÜ, Demirel C, Kilciksiz SC, Gürgül S, Zincircioğlu SB, Erdal N. Melatonin can Ameliorate Radiation-Induced Oxidative Stress and InflammationRelated Deterioration of Bone Quality in Rat Femur. Inflammation. 2016;39(3):1134-40. doi: 10.1007/ s10753-016-0347-x 61. Cakmak I, Simsek G, Yildiz A, Vardi N, Polat A, Tanbek K, Gurocak S, et al. Melatonin’s protective effect on the salivary gland against ionized radiation damage in rats. J Oral Pathol Med. 2016. doi: 10.1111/jop.12386 62. Koc M, Buyukokuroglu M, Taysi S. The effect of melatonin on peripheral blood cells during total body irradiation in rats. Biol Pharm Bull. 2002;25(5):656-7. doi: 10.1248/bpb.25.656 63. Cagnoli C, Atabay C, Kharlamova E, Manev H. Melatonin protects neurons from singlet oxygeninduced apoptosis. J Pineal Res. 1995;18(4):222-6. doi:

10.1111/j.1600-079X.1995.tb00163.x 64. Siu A, Reiter R, To C. Pineal indole amines and vitamin E reduce nitric oxide-induced lipid peroxidation in rat retinal homogenates. J Pineal Res.1999; 27(2): 122–128. doi: 10.1111/j.1600-079X.1999.tb00606.x 65. Sainz R, Mayo J, Uria H. The pineal neuro hormone melatonin prevents in vivo and in vitro apoptosis in thymocytes. J Pineal Res. 1995;19(4):178-88. doi: 10.1111/j.1600-079X.1995.tb00187.x 66. Maestroni G, Covacci V, Conti A. Hematopoietic rescue via T-cell-dependent, endogenous granulocyte macrophage colony-stimulating factor induced by the pineal neuro hormone melatonin in tumor-bearing mice. Cancer Rese. 1994;54(9): 2429-32. 67. Ruifrok A, Weil M, Thames H, Mason K. Diurnal variations in the expression of radiation-induced apoptosis. Radiat Res. 1998;149(4):360-5. doi: 10.2307/3579698 68. Shirazi A, Haddadi G, Minaee B. Evaluation of melatonin for modulation of apoptosis-related genes in irradiated cervical spinal cord. International Journal of Low Radiation. 2010; 7(6): 436–445. doi: 10.1504/ IJLR.2010.037665 69. Sharma S, Haldar C, Chaube S, Laxmi T, Singh S. Longterm melatonin administration attenuates low-LET -radiation- induced lymphatic tissue injury during the reproductive ly active and inactive phases of Indian palm squirrels (Funambulus pennanti). Br J Radiol. 2010;83(986):137-51. doi: 10.1259/bjr/73791461 70. Najafi M, Shirazi A, Motevaseli E, Rezaeyan AH, Salajegheh A, Rezapoor S. Melatonin as an anti-inflammatory agent in radiotherapy. Inflammopharmacology. 2017;25 (4):403-413. doi: 10.1007/s10787-017-0332-5 71. Hu ZP, Fang XL, Fang N, Wang XB, Qian HY, Cao Z, et al. Melatonin ameliorates vascular endothelial dysfunction, inflammation, and atherosclerosis by suppressing the TLR4/ NF-jB system in high-fat-fed rabbits. J Pineal Res. 2013; 55(4):388–398. doi: 10.1111/jpi.12085 72. Bertuglia S, Marchiafava PL, Colantuoni A Melatonin prevents ischemia reperfusion injury in hamster cheek pouch microcirculation. Cardiovasc Res. 1996; 31(6):947–952. doi: 10.1016/S0008-6363(96)00030-2 73. Sewerynek E, Reiter RJ, Melchiorri D, Ortiz GG, Lewinski A. Oxidative damage in the liver induced by ischemia-reperfusion: protection by melatonin. Hepatogastroenterology. 1995; 43(10):898-905. 74. Wu CC, Lu KC, Lin GJ, Hsieh HY, Chu P, Lin SH, et al. Melatonin enhances endogenous heme oxygenase‐1 and represses immune responses to ameliorate experimental murine membranous nephropathy. J Pineal Res. 2012; 52(4):460–469. doi: 10.1111/j.1600-079X.2011.00960.x 75. Maestroni GJ T‐Helper‐2 lymphocytes as a peripheral target of melatonin. J Pineal Res. 1995;18(2):84–89. doi: 10.1111/j.1600-079X.1995.tb00144.x 76. Bondy SC, Lahiri DK, Perreau VM, Sharman KZ, Campbell A, Zhou J, et al Retardation of brain aging by chronic treatment with melatonin. Ann N YAcad Sci. 2004;1035(1):197-215. doi: 10.1196/annals.1332.013 77. Jang SS, Kim HG, Lee JS, Han JM, Park HJ, Huh GJ et al. Journal of Radiobiology, Vol. 5, No. 1, March 2018

7

Guevara et al

Melatonin reduces X-ray radiation-induced lung injury in mice by modulating oxidative stress and cytokine expression. Int J Radiat Biol. 2013;89(2):97-105. doi: 10.3109/09553002.2013.734943 78. Michaeloudes C, Sukkar MB, Khorasani NM, Bhavsar PK, Chung KF. TGF-β regulates Nox4, MnSOD and catalase expression, and IL-6 release in airway smooth muscle cells. Am J Phys Lung Cell Mol Phys. 2011;300(2):L295-304. doi: 10.1152/ajplung.00134.2010 79. Chávez E, Reyes‐Gordillo K, Segovia J, Shibayama M, Tsutsumi V, Vergara P, et al. Resveratrol prevents fibrosis, NF‐κB activation and TGF‐β increases induced by chronic CCl4 treatment in rats. J Appl Toxicol.

2008;28(1):35–43. doi: 10.1002/jat.1249 80. Srinivasan V, Spence D, Pandi-Perumal S. Melatonin, environmental light, and breast cancer. Breast Cancer Res Treat. 2008;108(3):339-350. doi: 10.1007/s10549007-9617-5 81. Cui P, Luo Z, Zhang H. Effect and mechanism of melatonin’s action on the proliferation of human umbilical vein endothelial cells. J Pineal Res. 2006;41(4): 358–362. doi: 10.1111/j.1600-079X.2006.00375.x 82. Bhatia L. Melatonin: anti-aging perception and current perspectives. Aging Interventions and Therapies. 2005;147–169. doi: 10.1142/9789812701329_0008

Please cite this paper as: Guevara C, Herrera L, Cárdenas R, Mestre F, Suarez-Roca H. Biology of radiation-induced mandibular necrosis and the therapeutical perspectives for melatonin. J Radiobiol. 2018;5(1):e02. doi: 10.15171/ jrb.2018.02. Copyright © 2018 The Author(s); Published by Nickan Research Institute. This is an open-access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

8

Journal of Radiobiology, Vol. 5, No. 1, March 2018