Please cite this paper as:

Pal, P.K., Chattopadhyay, A. and Bandyopadhyay, D. 2020. Melatonin as a potential therapeutic molecule against COVID-19 associated gastrointestinal complications: An unrevealed link. Melatonin Research. 3, 3 (Jun. 2020), 417-435. DOI:https://doi.org/https://doi.org/10.32794/mr11250070.


Review

Melatonin as a potential therapeutic molecule against COVID-19 associated gastrointestinal complications: An unrevealed link

Palash Kumar Pal1, Aindrila Chattopadhyay2, Debasish Bandyopadhyay1*

1Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata-700009

2Department of Physiology, Vidyasagar College,39, Sankar Ghosh Lane, Kolkata-700006

*Correspondence:debasish63@gmail.com, Tel: +91 9433072066

Running titleCOVID-19 and gastrointestinal melatonin

Received: May 01, 2020; Accepted: June 12, 2020

 

ABSTRACT

     Outbreak of the novel coronavirus disease (COVID-19) was first reported in Wuhan, Hubei province of China, in early December 2019 which was later declared as a pandemic by World Health Organization (WHO) in March 2020. The International Committee on Taxonomy of Viruses has termed this novel coronavirus as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). According to the report of WHO on 29th April, 2020, 3018681 confirmed cases along with 207973 deaths have been documented globally. COVID-19 was originally reported as a lethal lung disease with fever and cough as the most common symptoms; however, the increasing number of gastrointestinal symptoms, such as diarrhoea, vomiting and abdominal pain in patients have clearly suggested that gastrointestinal tract (GIT) may also serve as a potential route for SARS-CoV-2 infection. To identify the effective therapies on this pandemic is urgent. Keeping this in mind, we realize that melatonin is a potent antioxidant, anti-inflammatory and immunomodulatory molecule and it has been used in diverse diseases and pathophysiological conditions, including respiratory disease and viral infections. Importantly, melatonin specific receptors and its endogenously synthetic machinery are distributed throughout the mammalian gastrointestinal system. Therefore, the therapeutic potentiality of melatonin in SARS-CoV-2 associated digestive symptoms cannot be ignored. In this review, we focus on the clinical implications of melatonin on the digestive complications associated with SARS-CoV-2 infection.

Key words: COVID-19, SARS-CoV-2, digestive complications, melatonin, gastrointestinal tract, inflammation, antioxidant, immunoregulator.

__________________________________________________________________________________________

 

1. INTRODUCTION

     Emergence of a novel coronavirus, belonging to the family coronaviride, pneumonia was first reported in Wuhan, Hubei province of China, in early December 2019 (1-3). Following virus isolation and identification from human airway epithelial cells, International Committee on Taxonomy of Viruses (ICTV) has designated this novel coronavirus as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on 7th January, 2020 (1, 4, 5).  World Health Organization (WHO) named this pneumonia as coronavirus disease 2019 (COVID-19) on 11th February, 2020 (2) and declared it as a pandemic on 11th March, 2020. Although most of the coronavirus are harmless to human but beta coronaviruses, SARS-CoV (6) and Middle East respiratory syndrome coronavirus (MERS-CoV) (7) repeatedly cause lethal respiratory diseases in humans and their non-segmented positive sense RNA are mostly distributed in diverse mammals, including human (1, 8, 9). Till to 29th April, 2020, globally 3018681 confirmed cases along with 207973 deaths have been documented by WHO. Despite of the fact that COVID-19 was reported as a lethal lung disease with fever and cough as the most common symptoms (9, 10), the increasing number of gastrointestinal symptoms, such as diarrhoea, vomiting and abdominal pain in  patients (2-10%) have implicated that SARS-CoV-2 might also utilize the digestive systems as a route of infection (9-12). This potential GI route infection has been masked by the profound respiratory symptoms occurred in the COVID-19 patients until to a single-cell transcriptomic study which has demonstrated this route of SARS-CoV-2 infection (13-14).

Melatonin (N-acetyl-5-methoxytryptamine), a tryptophan derivative, is well known for its multiple physiological activities and it has been used to treat diseases and pathophysiological conditions related to virus infections, particularly to virus associated respiratory diseases (15-18). Therefore, under the current pandemic situation, testing of the potential effects of melatonin on respiratory and other symptoms associated to SARS-CoV-2 is encouraged. Interestingly, melatonin binding sites or its receptors and its endogenously synthetic machinery are distributed throughout the entire mammalian GIT (19-20). This suggests the local actions of melatonin as a potent anti-oxidant, anti-inflammatory and immuno-modulatory molecule (18, 21-22). Herein, based on the available information we hypothesize that melatonin, especially the gastrointestinal original one, may play a critical role in the prevention and treatment of SARS-CoV-2 associated gastrointestinal symptoms. Under current situation, this hypothesis warrants careful examination.

 

2. GASTROINTESTINAL TRACT: A POTENT TARGET OF SARS-CoV-2

     Based on the clinical evidence gathered till date, it is clear that although SARS-CoV-2 primarily affects the respiratory system but its pathogenicity is not confined to the lungs only, rather it also damages other vital organs in critically illed patients leading to multiple organ dysfunction syndromes (23-25). Usually, coronavirus attacks the upper respiratory, gastrointestinal and central nervous systems in mammals (26). Specific to SARS-CoV-2, the gastrointestinal tract is also its common route for invasion. This has been confirmed in afflicted patients with digestive symptoms including, most commonly, anorexia and diarrhoea (3, 13, 27, 28).

     Intestinal epithelial cells serve as a potent barrier in protecting against microbial infection by initiating immune responses (29). Viruses, such as coronavirus, rotavirus and noroviruses, prefer to attack the absorptive enterocytes and damage the normal absorption process that causes imbalance in intestinal secretion and activates enteric nervous system (30-32). This has been observed also in SARS-CoV-2 infection since this virus is detected in the oesophagus, stomach, duodenum and rectum, even in the faecal samples (52.4%) of COVID-19 patients (3). Moreover, detection of SARS-CoV-2 RNA in the stool of COVID-19 patients further confirms this contention (28, 33-35). The presence of viral replication in the susceptible intestinal cells, particularly in angiotensin converting enzyme-2 (ACE2)- and transmembrane protease serine 2 (TMPRSS2)-expressing oesophageal upper cells and absorptive enterocytes leads to GI symptoms (diarrhoea and nausea) 24-48 hrs prior to the appearance of fever and respiratory symptoms in some patients (14, 36, 37). In severe cases, SARS-CoV-2 even binds and manipulates the expression of ACE2 in hepatic tissue causing liver injuries (11). On the other hand, SARS-CoV-2 can modify the performance of microflora in GIT and such alterations can interfere respiratory system and vice versa (via “gut-lung axis”), possibly through the mucosal immune system (38, 39). Collectively, the increasing GI symptoms in COVID-19 patients probably indicates profound elevation in viral load as well as their replication in the GI cells which eventually increases the disease severity (36).

 

3. TRANSMISSION OF SARS-CoV-2 IN THE GASTROINTESTINAL TRACT

     In general, direct contact or air droplets are the principal route of SARS-CoV-2’s human-to-human transmission (1, 2, 37) since person’s contact within the distance of 1 meter from each other shows the maximum risk of SARS-CoV-2 infection (40). Although the exact pathway or route through which SARS-CoV-2 infects the GIT is still not clarified, some clues are present. The co-expression pattern analysis of single-cell RNA sequencing data indicates that the SARS-CoV-2 virus enters into the host intestinal cells by interacting with ACE2 and TMPRSS2 as the receptors which are located in the cell membrane. The cells that express both the receptors are the most vulnerable for SARS-CoV-2 infection, while cells which only express one of the receptors are more resistance to this virus attack (14, 41-43). For example, COVID-19 patients usually have elevated co-expression of ACE2 and TMPRSS2 in the oesophageal upper epithelial and gland cells, and absorptive enterocytes of the ileum and colon (14, 42). These observations addressed the importance of both ACE2 and TMPRSS2 receptors in the pathogenesis of COVID-19 (43). It reveals, for the first time, the possibility of extra-pulmonary spread of SARS-CoV-2 in the infected individuals (14, 42). In addition, the virulence of SARS is also associated with increased permeability to intestinal lipopolysaccharide and bacterial transmigration through the gastrointestinal wall (44).

     Transmissibility and pathogenesis of coronavirus in human are  primarily a resultant of an interaction cascade, i.e., - virus first attaches to the host cell followed by recognition of specific receptor, then cleavage by protease and fusion with membrane, finally its transmembrane spike glycoprotein (S-protein) binds to cell specific receptor (ACE2 and TMPRSS) -binding domain and enters into cells (12). ACE2 is a zinc metallopeptidase that converts angiotensin II to angiotensin I-VII (1, 12, 14, 43). The property of ACE2 to interact with a predetermined receptor-binding domain of CTD1 in SARS-CoV promotes its efficiency in spreading the infection among species, thus increasing person-to-person transmission (45, 46). Different configurations (‘up’ and ‘down’) of CTD1 facilitate binding to ACE2 by controlling the interaction between CTD1, CTD2, S1-ACE2 complex and the S2 subunit (47). Structural analysis of SARS-CoV-2 S protein revealed its higher binding affinity to ACE2 than that of SARS CoV S protein (37, 48, 49). On the other hand, cleavage of SARS-S protein by TMPRSS2 promotes SARS-CoV-2 entering susceptible cells in an endosomal pathway independent manner, thus facilitating the virus to release fusion peptide for membrane fusion (50). Thereafter, cleavage of ACE2 increases the viral infectivity by about 30-folds, since the cleavage of SARS-CoV- 2 spike protein possesses higher furin score (0.688) when compared to SARS-CoV (0.139) (27, 51-53).

     Immune system plays a pivotal role in the pathogenesis of COVID-19. During the preliminary stages of infection, SARS-CoV-2 activates the dendritic and epithelial cells, and  triggers release of numerous pro-inflammatory cytokines and chemokines including IL-1β, IL-2, IL-6, IL-8,  interferons (IFN-α/β), tumor necrosis factor (TNF), CeC motif chemokine 3 (CCL3), CCL5, CCL2, IP-10, etc. All these immune events collectively promote uncontrolled generation of these cytokines and chemokines referred to as cytokine storm, eventually leading to severe COVID-19 disease state (54-56).


4. ACE2 BASED STRATEGIES TO COMBAT COVID-19

     The molecular mechanisms of COVID-19 pathogenesis in the gastrointestinal tract remain most elusive; however, several ACE2 based treatment strategies have been proposed. For example, ACE2 fusion proteins and TMPRSS2 inhibitors are recommended for rapid diagnosis or, treatment of COVID-19 (2). ACE2 inhibitors are also considered since GL1001 (selective ACE2 inhibitor) was demonstrated to reduce DSS-induced mucosal inflammation and distal colon pathology in inflammatory bowel disease (IBD) animal model (57). In addition, ACE2 inhibition is thought to elevate its substrate level which in turn would block over production of different pro-inflammatory cytokines (58). Noteworthy, SARS-CoV-2 is known to trigger cytokine storm, leading to multi-organ dysfunction in severe COVID-19 pneumonia patients (12). Based on these observations, it may be assumed that SARS-CoV-2 interacts with the gastrointestinal ACE2 receptor to increase mucosal membrane permeability, promoting generation of inflammatory cytokines and eventually leading to the destructive cytokine storm status (10). Thus, use of ACE2 inhibitors to reduce or inhibit SARS-CoV-2 induced gastrointestinal symptoms has future therapeutic relevance (10). The compounds which possess both anti-inflammatory and immunomodulatory properties seem to deliver positive effects in preventing GIT against such cytokine storm. In this regard, melatonin appears to be such a compound and its anti-inflammatory and immunomodulatory activities have been frequently reported in numerous animal and human studies (18, 59-62).

 

5. MELATONIN: A POTENTIAL REMEDY FOR COVID-19 ASSOCIATED GI

    PATHOLOGY

     Melatonin can easily exert its actions on the targeting cells/organs since it is permeable to any cell or cell compartment due to its amphipathic nature. Moreover, presence of melatonin specific membrane transporters in the cell and mitochondrial membranes provides an advantage to move this molecule against its concentration gradient and accumulated in important organelles such as in mitochondria (63-64). The widely distributed melatonin receptors in diverse mammalian tissues, including the GIT, suggest its pleiotropic activities (21, 22, 65-67). Among these multiple activities, the antioxidant, anti-inflammatory and immunomodulatory effects have been well documented (21, 68-71). Even though lack of report on its direct viricidal action, the indirect anti-viral property of melatonin has been suggested (18). Several studies have shown that melatonin decreases the virus load (72), prevents oxidative stress mediated acute lung injury and inhibits unwanted immune response in mammals (18). The combination of melatonin’s activities including potent antioxidant, anti-inflammatory, immunomodulatory and indirect anti-viral action are warranted to test therapeutic effects of this molecule on the digestive complications caused by SARS-CoV-2 infection.

5.1. Effects of melatonin in the gastrointestinal tract.

     Identification of immunolocalization of melatonin in the enterochromaffin cells (EC) of mammalian digestive mucosa (73) and detection of the rate limiting enzymes of melatonin biosynthesis- arylalkylamine-N-acetyltransferase (AANAT) (21, 74-75) and hydroxyindole-O-methyltransferase (HIOMT) (76) confirm the synthesis of melatonin in the gastrointestinal tract of mammals. Further identification of melatonin specific membrane receptors and binding sites in the mammalian GIT uncovers the paracrine actions of melatonin (19, 77, 78). The locally synthesized melatonin primarily binds to its G-protein coupled membrane receptors- MT1 and MT2 to exert its paracrine activities while another receptor MT3 (human quinone reductase 2) is also demonstrated to be involved in some actions of melatonin (79-80). Melatonin plays the critical roles under oxidative stress or inflammation receptor-independently or receptor-dependently to regulate intracellular signalling proteins, particularly antioxidant and pro-oxidant enzymes in GIT (66-67, 77, 79).

     Being a well-known antioxidant, melatonin scavenges extremely toxic peroxynitrite anion, hydroxyl-, peroxynitrite-, peroxyyl radicals and singlet oxygen (81-83), thus it inhibits peroxidation of membrane lipids (84) and reduces the severity of inflammation (85, 86). Such direct antioxidant and free radical scavenging activities of melatonin are well examined in numerous in vitro and in vivo studies (85, 87, 88). Moreover, interaction of melatonin with reactive oxygen species (ROS) generates several metabolites which also have potent antioxidant property similar to melatonin (81, 86, 89). Notably, viral infections and their replication undergo spontaneous oxidation and provoke ROS formation Similarly, SARS-CoV-2 infection also causes generation of excessive ROS (14, 90). Thus, in the case of COVID-19 disease, melatonin is an ideal antioxidant to reduce the oxidative damage caused by SARS-CoV-2 infection.

5.2. Effects of melatonin on the microbiota of GIT and its association with covid-19.

     Distribution of the normal microbial population in the GIT is essential to maintain the health and function of the intestine. Disturbance in such distribution and microbial signalling leads to gastrointestinal complications, known as “gut dysbiosis”- a phenomenon associated with numerous gastrointestinal diseases and disorders (91, 92). Similar to the gut micro environment, lungs also possess its own distinct microorganism population (93). Bacteroidetes and Firmicutes are abundant in the GIT, while they are also the predominating ones in the lungs (94). The gut microbes can modulate the pulmonary health through a bidirectional cross-talk termed as “gut-lung axis” (95). Under pathological condition, excessive endotoxins and microbial metabolites of gastrointestinal origin are released into the blood stream to cause inflammation in the lungs and vice-versa (96). It can be assumed that some gastrointestinal complications are caused by disruption of the gut micro environment after SARS-Cov2 infection. Gut microbes are reported to play a potent role in the pathogenesis of acute respiratory distress syndrome (ARDS) (97)- one of the major symptoms of SARS-Cov2 infection. On the other hand, the microbiota is responsible for the innate and adaptive immune responses of the GIT (98). In order to maintain the gut homeostasis, the pro-inflammatory and anti-inflammatory system must be balanced and such balance is performed by the commensal microorganisms (99). Usually, this homeostasis is disturbed by virus infections such as observed in the SARS-Cov2 invasion (100-103) and causes gastrointestinal symptoms in COVID-19 patients (101, 103-105).

     In this context, melatonin is capable of maintaining the health of the intestine (106). Melatonin supplementation prevents the metabolic disturbances in varied animal studies possibly through re-establishment of the balance of beneficial microbiota in gut (107, 108). Melatonin also reduces gut membrane permeability and suppresses immune response initiated by Escherichia coli in gut (109). The importance of melatonin and its local production in the protection of GIT has been reported by Chojnacki et al. (110). They found that Helicobacter pylori infection downregulated the expression of AANAT and HIOMT, thus decreasing the endogenous synthesis of melatonin in the GIT while the melatonergic system was restored to normal after the infection abolishes. In light of the discussion, use of melatonin may help to rebuild the disturbed GIT microtia caused by SARS-Cov2 infection and improve the GI symptoms in COVID-19 patients.

5.3. Anti-inflammatory and immuno-modulatory properties of melatonin: an armament against covid-19 induced gastrointestinal complications.

     Accompanied with its antioxidant property, melatonin also possesses potent anti-inflammatory and immunomodulatory actions in diverse mammalian tissues, particularly in the gastrointestinal tract (18, 21, 69, 111-113). For example, transcription factor, NF-kβ is extremely sensitive to oxidative stress and under this stress it triggers expression of different pro-inflammatory genes and increases the state of inflammation in cells (69, 114). By suppressing oxidative stress melatonin overcomes inflammatory response primarily via modulation of NF-kβ associated signalling pathways involving TLR4 and TLR5 (68-69, 115). Melatonin downregulates the expression of TLR4 and other genes associated with its signalling cascade including MyD88 (116). In most of the cases, melatonin inhibits NF-kβ expression by increasing Ikβ level which suppresses the excessive production of adhesive agents, leukocytes and other inflammatory cells (117), ultimately restricting the level of inflammatory reactions in the GI tissue. Similarly, melatonin also inhibits the level of proinflammatory cytokine IL-1β in aflatoxin B1 induced rat intestinal lesions (118). Mast cells are important innate immune cells in GIT and abundance of melatonin receptors are present in these cells. By acting on these receptors, melatonin restricts the release of TNF-α from mast cells (119) and inhibits the activation of mast cells by down-regulating the expression of the NF-κB to exert its anti-inflammatory and immunoregulatory activities (120). The other anti-inflammatory pathways of melatonin include inhibition of the production of prostaglandins and adhesion molecules (85), restricting leukocyte transendothelial cell migration (121), cyclooxygenase 2 expression in the macrophages (122) and inhibiting the recruitment of various pro-inflammatory cells to the inflammation site (85, 123). Interestingly, AFMK and AMK- the metabolites of melatonin also play potent anti-inflammatory actions as does melatonin (123).

      It is noted that the overreaction of the innate immune responses and excessive inflammatory reaction are the major risk factors associated to the severity and mortality of COVID-19 patients (9-11, 14, 71, 124-126). This provides the rationality behind the use of melatonin in preventing and treating gastrointestinal complications caused by such overreaction of the innate immune response and excessive inflammatory reaction in COVID-19 patients. As mentioned earlier, SARS-CoV-2 triggers the dendritic cells and epithelial cells to release pro-inflammatory cytokines and chemokines (Figure 1) creating a destructive cytokine storm and eventually increasing the disease severity (54-56). A profound reduction in the circulating levels of neutrophils and lymphocytes was also noticed in COVID-19 patients (10, 127). Although endoscopic studies did not reveal any macroscopic inflammation in the GIT, but numerous infiltrating plasma cells and lymphocytes with interstitial oedema were noted in the lamina propria of the stomach, duodenum and rectum of a COVID-19 patient clearly suggesting an activation of mucosal immune cells (128). Further study on the inflammatory markers in affected patients indicated profound increase in their circulatory levels of procalcitonin, C reactive protein, D-dimer and ferritin (5,124); and TNF, IL-2R, IL-6, IL-8 and IL-10 levels were also elevated in the serum of patients with severe infection, but levels of IL-1β and interferon-gamma producing CD4+ and CD8+ T cells were not affected (128). However, plasma levels of IL-1beta, IL-1ra, IL-2, IL-4, IL-6, IL-7, IL-8, IL-9, IL-10, IL-13, IL-17, fibroblast growth factor, granulocyte colony-stimulating factor, granulocyte macrophage-colony stimulating factor, interferon-gamma, TNF, vascular endothelial growth factor, IP-10, monocyte chemoattractant protein (MCP)-1, macrophage inflammatory protein (MIP)-1alpha, platelet-derived growth factor and MIP-1beta were distinctly increased in ICU patients. Collectively, the over responses of the immune cells to SARS-CoV-2 invasion caused cytokine storm syndrome in COVID-19 patients (1, 125).

     The evidence mentioned above suggests the importance of the innate immune system in GIT for SARS-CoV-2 infection. In the GIT, intestinal mucosa serves as the primary defensive layer against intruded pathogens. Apart from its basic absorptive enterocytes, goblet cells, enteroendocrine cells, and Paneth cells, the underlying lamina propria exclusively comprises of the immunocompetent cells that are responsible for the innate immune response in the mucosa (126). Pathogens often bind to pathogen-recognition receptors (PRRs), most often the TLRs, localized on the cell surface or endosomes of the intestinal epithelial cells and other immunocompetent cells in the lamina propria to initiate innate immune responses (126). Other PRRs including Nucleotide-binding oligomerization domain (NOD) (129-130) and nucleotide-binding domain leucine-rich repeat-containing receptors (NLRs) (131-132) are also responsible for pathogen innate immune responses and inflammation. NLPR3 inflammasome represents the most common inflammatory reaction involving ASC, caspase-1, a pro-IL-1β and pro-IL-18 to form mature and secreted interleukins (126). Gut-associated lymphoid tissue (GALT) regulates IgA production and its release from the intestinal B cells into the lumen to maintain the intestinal homeostasis (126). Paneth cells, the prime source of antimicrobial proteins, synthesize and secrete lysozyme, secretory phospholipase A2, C-lectin RegIIIγ, α-defensins and angiogenin-4 to clean the pathogen (126). All these innate immune responses can be activated by SARS-Cov-2 infection and the uncontrolled innate immune responses often occur in the severe COVID-19 patients with intestinal symptoms (2, 14, 133). The capability of SARS-CoV to deplete mucosal lymphoid tissue in the pharynx, appendix, and small intestines has been reported in humans (134) and the similar reactions are expected to occur also in SARS-CoV-2 infection. The SARS-CoV positive in situ hybridization signals were also identified in the cytoplasm of mucosal epithelial cells (135, 136) as well as in mucosal and submucosal lymphocytes (136). Electron microscopic studies revealed the abundance of viral particles in the mucosal epithelial cells, particularly in the dilated endoplasmic reticulum and surface of the microvilli (136-137). Viral proteins and genomic sequences were also detected in the gastric parietal cells (138).

Melatonin seems to be of utmost importance to control the unbalanced innate immune response in GIT induced by SARS-CoV-2 infection. Animal studies have unveiled the potent roles of melatonin in the regulation of the immune response under inflammatory states in the bone marrow and other tissues where the proliferation and maturation of the lymphocytes (both T and B), NK cells, granulocytes and monocytes occur (139). Furthermore, melatonin treatment improves antigen presentation in macrophages possibly through induction of complement receptor 3, MHC class I and class II, and CD4 antigens (140). Melatonin treatment also downregulates the circulating levels of different proinflammatory interleukins (IL-1β, IL-6, IL-17), interferon-γ, TNF-α, kinase Cζ (PKCζ) and calmodulin 3 (CALM3) (66, 141-143). The damages in the colonic mucosa and other parts of the GIT related to autoimmune response, inflammation and sepsis are significantly reduced with melatonin treatment (144, 145). Clinical trials also show that melatonin supplementation reduces the circulating levels of TNF-α and IL-6 of patients (146). In acute lung injury, similar to found in COVID-19 disease, the anti-inflammatory and immunomodulatory actions of melatonin play important roles to protect against this injury (15) (Figure 1). Thus, melatonin is a potent compound with antioxidant, anti-inflammatory and immunomodulatory properties- the three essential characteristics in a single compound to combat COVID-19 induced digestive disorders. 

Figure 1.jpg

Fig. 1. Hypothetical model of the protective role of melatonin against SARS-CoV-2 induced gastrointestinal inflammation.

     Blue lines indicate SARS-CoV-2 induced inflammatory pathways, while green lines indicate melatonin mediated pathways of protection. MTR- Melatonin receptor; SARS-CoV-2- severe acute respiratory syndrome coronavirus 2.

 

6. FUTURE PERSPECTIVES

     Finally, it is essential to know about the safety measures of this molecule. In this regard, melatonin is ideal since its short-term and long-term use in human, even in ICU patients, reported no serious adverse effects (147-149). Similarly, melatonin at higher dose (1 g/day) for prolonged period use in human, or in acute lung injury animal models has not shown any adverse effect (17, 18, 150, 151). Since the effect of melatonin has not been tested in COVID-19 patient in a large scale, verification of the therapeutic effects of melatonin in COVID-19 patients is recommended (133). Melatonin has the capacity to inhibit exaggerated cytokine storm in the infected individuals, thus it can restrict the severity of COVID-19 disease (152, 153), particularly in the gastrointestinal tract. Collectively, in light of these evidences it is likely that melatonin has the positive effects on digestive complications in COVID-19 patients. Its beneficial effects may be superior to other contemporary antioxidant molecules. However, such hypothesis is worth with the extensive clinical trials in the future.


ACKNOWLEDGEMENTS

     Dr. Palash Kumar Pal gratefully acknowledges the receipt of UGC Dr. D. S. Kothari Post Doctoral Fellowship (BL/16-17/0502), Govt. of India. Dr. Aindrila Chattopadhyay is supported by funds available to her from Department of Science and Technology, Govt. of West Bengal. Prof. DB is also supported from departmental BI grant of University of Calcutta. DB also gratefully acknowledges the support he received from DST-PURSE Program awarded to University of Calcutta. Prof. DB gratefully acknowledges the contribution of the Editor-In-Chief of Melatonin Research, Dr. Dun-Xian Tan in critically reading and editing of the manuscript which has definitely improved the scientific and readership quality of the article.


AUTHORSHIP

     Prof. DB contributed to conception, revised the manuscript and approved it, Dr. AC critically read and revised the manuscript and approved it. Dr. PKP also contributed to conception, prepared figures, drafted the manuscript and incorporated corrections into the manuscript as and when required.


CONFLICT OF INTEREST

     Authors declare no conflict of interest.

 

REFERENCES

 

  1. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W(2020) A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 382: 727–733. DOI: 10.1056/NEJMoa2001017.

  2. Gu J, Han B, Wang J (2020) COVID-19: gastrointestinal manifestations and potential fecal-oral transmission. Gastroenterology S0016-5085(20)30281-X.

  3. Ng SC, Tilg H (2020) COVID-19 and the gastrointestinal tract: more than meets the eye. Gut. pii: gutjnl-2020-321195. DOI: 10.1136/gutjnl-2020-321195.

  4. Phelan AL, Katz R, Gostin LO (2020) The novel coronavirus originating in Wuhan, China: Challenges for global health governance. JAMA DOI: 10.1001/jama.2020.1097.

  5. Wu Y, Guo C, Tang L, Hong Z, Zhou J, Dong X, Yin H, Xiao Q, Tang Y, Qu X, Kuang L, Fang X, Mishra N, Lu J, Shan H, Jiang G, Huang X (2020) Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol. Hepatol. 5: 434-435. DOI: 10.1016/S2468-1253(20)30083-2.

  6. Kuiken T, Fouchier RA, Schutten M, Rimmelzwaan GF, van Amerongen G, van Riel D, Laman JD, de Jong T, van Doornum G, Lim W, Ling AE, Chan PK, Tam JS, Zambon MC, Gopal R, Drosten C, van der Werf S, Escriou N, Manuguerra JC, Stöhr K, Peiris JS, Osterhaus AD (2003) Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome.  Lancet 362: 263-270. DOI: 10.1016/S0140-6736(03)13967-0.

  7. de Groot RJ, Baker SC, Baric RS, Brown CS, Drosten C, Enjuanes L, Fouchier RA, Galiano M, Gorbalenya AE, Memish ZA, Perlman S, Poon LL, Snijder EJ, Stephens GM, Woo PC, Zaki AM, Zambon M, Ziebuhr J (2013) Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Coronavirus Study Group. J. Virol .87: 7790-7792. DOI: 10.1128/JVI.01244-13.

  8. Su S, Wong G, Shi W, Liu J, Lai ACK, Zhou J, Liu W, Bi Y, Gao GF (2016) Epidemiology, Genetic Recombination, and Pathogenesis of Coronaviruses. Trends Microbiol24: 490-502. DOI: 10.1016/j.tim.2016.03.003.

  9. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu  J, Gu X,  Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B (2020) Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China, Lancet 395: 497–506. https://doi.org/10.1016/S0140-6736(20)30183-5.

  10. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, Qiu Y, Wang J, Liu Y, Wei Y, Xia J, Yu T, Zhang X, Zhang L (2020) Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 395: 507-513. DOI: 10.1016/S0140-6736(20)30211-7.

  11. Guan W, Ni Z, Hu Y, et al. (2020) Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 382: 1708-1720. DOI: 10.1056/NEJMoa2002032.

  12. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, et al. (2020) Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395: 497-506. DOI: 10.1016/S0140-6736(20)30183-5.

  13. Leung JM, Yang CX, Tam Anthony, Shaipanich T, Hackett TL, Singhera GK, Dorscheid DR, Sin DD (2020) ACE-2 expression in the small airway epithelia of smokers and COPD patients: Implications of COVID-19. Eur. Respir. J. DOI: 10.1183/13993003. 00688-2020.

  14. Zhang H, Kang Z, Gong H, Xu D, Wang J, Li Z, Li Z, Cui X, Xiao J, Zhan J, Meng T, Zhou W, Liu J, Xu H (2020) Digestive system is a potential route of COVID-19: an analysis of single-cell coexpression pattern of key proteins in viral entry process. Gut 0: 1–9. DOI: 10.1136/gutjnl-2020-320953.

  15. Huang SH, Cao XJ, Liu W, Shi XY, Wei W (2010) Inhibitory effect of melatonin on lung oxidative stress induced by respiratory syncytial virus infection in mice. J. Pineal Res. 48: 109–116. https://doi.org/10.1111/j.1600-079X.2009.00733.x.

  16. Yip HK, Chang YC, Wallace CG, Chang LT, Tsai TH, Chen YL, Chang HW, Leu S, Zhen YY, Tsai CY, Yeh KH, Sun CK, Yen CH (2013) Melatonin treatment improves adipose-derived mesenchymal stem cell therapy for acute lung ischemia-reperfusion injury. J. Pineal Res. 54: 207–221. https://doi.org/10.1111/jpi.12020.

  17. Wu X, Ji H, Wang Y, Gu C, Gu W, Hu L, Zhu L (2019) Melatonin alleviates radiation- induced lung injury via regulation of miR-30e/NLRP3 axis, Oxidative Med. Cell. Longev. 2019: 4087298. https://doi.org/10.1155/2019/4087298.

  18. Reiter RJ, Ma Q, Sharma R (2020) Treatment of Ebola and other infectious diseases: melatonin “goes viral”. Melatonin Res. 3: 43–57. https://doi.org/10.32794/ mr11250047.

  19. Bubenik GA (1999) Localization and physiological significance of gastrointestinal melatonin. In: R Watson (ed.) Melatonin in health promotion. CRC press, Boca Raton, Florida, pp. 21–39.

  20. Kvetnoy IM, Ingel IE, Kvetnaia TV Malinovskaya NK, Rapoport SI, Raikhlin NT, Trofimov AV, Yuzhakov VV (2002) Gastrointestinal melatonin: cellular identification and biological role. Neuro. Endocrinol. Lett. 23: 121–132.

  21. Pal PK, Bhattacharjee B, Ghosh A, Chattopadhyay A, Bandyopadhyay D (2018) Adrenaline induced disruption of endogenous melatonergic system, antioxidant and inflammatory responses in the gastrointestinal tissues of male Wistar rat: an in vitro study. Melatonin Res.1: 109–131. DOI: 10.32794/mr11250007.

  22. Pal PK, Sarkar S, Chattopadhyay A, Tan DX, Bandyopadhyay D (2019) Enterochromaffin cells as the source of melatonin: key findings and functional relevance in mammals. Melatonin Res. (4): 61-82. DOI: https://doi.org/https://doi.org/10.32794 /mr11250041.

  23. Hu LL, Wang WJ, Zhu QJ, Yang L (2020) Novel coronavirus pneumonia related liver injury: etiological analysis and treatment strategy. Zhonghua Gan Zang Bing Za Zhi28: E001.

  24. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, Wu Y, Zhang L, Yu Z, Fang M, Yu T, Wang Y, Pan S, Zou X, Yuan S, Shang Y (2020) Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir. Med. DOI: 10.1016/S2213-2600(20)30079-5.

  25. Yao N, Wang SN, Lian JQ, Sun YT, Zhang GF, Kang WZ, Kang W (2020) Clinical characteristics and influencing factors of patients with novel coronavirus pneumonia combined with liver injury in Shaanxi region. Zhonghua Gan Zang Bing Za Zhi28: E003. DOI: 10.3760/cma.j.cn501113-20200226-00070.

  26. Perlman S, Netland J (2009) Coronaviruses post-SARS: update on replication and pathogenesis. Nat. Rev. Microbiol.7: 439-450.

  27. Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, et al., (2020) Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 395 (10224): 565-574. DOI: 10.1016/S0140-6736(20)30251-8.

  28. Mao R, Liang J, Shen J, Ghosh S, Zhu LR, Yang H, Wu KC, Chen MH (2020) Implications of COVID-19 for patients with pre-existing digestive diseases. Lancet 5: 426-428. https://doi.org/10.1016/S2468-1253(20)30076-5.

  29. Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, et al., (2017) A single-cell survey of the small intestinal epithelium. Nature 551 (7680): 333-339. DOI: 10.1038/nature24489.

  30. Crawford SE, Ramani S, Tate JE, Parashar UD, Svensson L, Hagbom M, Franco MA, Greenberg HB, O'Ryan M, Kang G, Desselberger U, Estes MK (2017) Rotavirus infection. Nat. Rev. Dis. Primers. 3: 17083. DOI: 10.1038/nrdp.2017.83.

  31. Ettayebi K, Crawford SE, Murakami K, Broughman JR, Karandikar U, Tenge VR, et al. (2016) Replication of human noroviruses in stem cell-derived human enteroids. Science 353: 1387–1393. DOI: 10.1126/science.aaf5211.

  32. Desmarets LMB, Theuns S, Roukaerts IDM, Acar DD, Nauwynck HJ (2014) Role of sialic acids in feline enteric coronavirus infections. J. Gen. Virol. 95: 1911–1918. DOI: 10.1099/vir.0.064717-0.

  33. Holshue ML, DeBolt C, Lindquist S, Lofy KH, Wiesman J, Bruce H, et al. (2020) First case of 2019 novel coronavirus in the United States. N. Engl. J. Med. 382: 929–936. DOI: 10.1056/NEJMoa2001191.

  34. Tang A, Tong ZD, Wang HL, Dai YX, Li KF, Liu JN, Wu WJ, Yuan C, Yu ML, Li P, Yan JB (2020) Detection of novel coronavirus by RT-PCR in stool specimen from asymptomatic child, China. Emerg. Infect. Dis.26 (6): DOI: 10.3201/eid2606.200301.

  35. Xie C, Jiang L, Huang G, Pu H, Gong B, Lin H et al. (2020) Comparison of different samples for 2019 novel coronavirus detection by nucleic acid amplification tests. Int. J. Infect. Dis. 93: 264-267. DOI: 10.1016/j.ijid.2020.02.050.

  36. Pan L, Mu M, Yang P, Sun Y, Wang R, Yan J, Li P, Hu B, Wang J, Hu C, Jin Y, Niu X, Ping R, Du Y, Li T, Xu G, Hu Q, Tu Lei (2020) Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China: a descriptive, cross-sectional, multicenter study. DOI: 10.14309/ajg.0000000000000620.

  37. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. (2020) Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. DOI: 10.1001/jama.2020.

  38. Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, Hansbro PM (2017) Emerging pathogenic links between microbiota and the gut-lung axis. Nat. Rev. Microbiol. 15: 55-63.

  39. He Y, Wen Q, Yao F, Xu D, Huang Y, Wang J (2017) Gut-lung axis: The microbial contributions and clinical implications. Crit. Rev. Microbiol. 43: 81-95.

  40. Repici A, Maselli R, Colombo M, Gabbiadini R, Spadaccini M, Anderloni A, Carrara S,  Fugazza A, DiLeo M, Galtieri PA, Pellegatta G, Ferrara EC, Azzolini E, Lagioia M (2020) Coronavirus (COVID-19) outbreak: what the department of endoscopy should know. Gastrointest. Endosc. 1-6. https://doi.org/10.1016/j.gie.2020.03.019.

  41. Chai X, Hu L, Zhang Y, Han W, Lu Z, Ke A, et al., (2020) Specific ACE2 expression in cholangiocytes may cause liver damage after 2019-nCoV infection. BioRxiv DOI: https://doi.org/10.1101/2020.02.03.931766.

  42. Liang W, Guan W, Chen R, Wang W, Li J, Xu K, Li C, Ai Q, Lu W, Liang H, Li S, He J (2020) Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol.21 (3): 335-337. DOI: 10.1016/S1470-2045(20)30096-6.

  43. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, et al., (2020) A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579:270–273. DOI: 10.1038/s41586-020-2012-7.

  44. Powers JH, Bacci ED, Guerrero ML, Leidy NK, Stringer S, Kim K, et al., (2018) Reliability, validity, and responsiveness of influenza patient-reported outcome (FLU-PRO©) scores in influenza-positive patients. Value Health. 21: 210–218. DOI: 10.1016/j.jval.2017.04.014.

  45. Gui M, Song W, Zhou H, Xu J, Chen S, Xiang Y, Wang X (2017) Cryo- electron microscopy structures of the SARS–coV spike glycoprotein reveal a prerequisite conformational state for receptor binding. Cell Res. 27: 119–129. https://doi.org/10.1038/cr.2016.152.

  46. Li et al., 2005 Li F, Li W, Farzan M (2005) Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 309: 1864–1868.

  47. Song W, Gui M, Wang X, Xiang Y (2018) Cryo- em structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog. 14: e1007236. DOI: 10.1371/journal.ppat.1007236.

  48. Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, Graham BS, McLellan JS (2020) Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367: 1260–1263. DOI: 10.1126/science.abb2507.

  49. Xu X, Chen P, Wang J, Feng J, Zhou H, Li X, Zhong W, Hao P (2020) Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission. Sci. China Life. Sci. 63: 457–460. DOI: 10.1007/s11427-020-1637-5.

  50. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al., (2020) SARS-coV-2 cell entry depends on ACE2 and TMPRSS 2 and is blocked by a clinically proven protease inhibitor. Cell 181 (2): 271-280.e8.DOI: 10.1016/j.cell.2020.02.052.

  51. Heurich A, Hofmann-Winkler H, Gierer S, Liepold T, Jahn O, Pöhlmann S (2014) Tmprss2 and ADAM17 cleave ace2 differentially and only proteolysis by TMPRSS 2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein. J. Virol. 88: 1293–1307. DOI: 10.1128/JVI.02202-13.

  52. Zhou Y, Vedantham P, Lu K, Agudelo J, Carrion R Jr, Nunneley JW, et al. (2015) Protease inhibitors targeting coronavirus and filovirus entry. Antiviral Res. 116: 76–84. DOI: 10.1016/j.antiviral.2015.01.011.

  53. Meng T, Cao H, Zhang H, Kang Z, Xu D, Gonh H, et al., (2020) The insert sequence in SARS- coV-2 enhances spike protein cleavage by TMPRSS. bioRxiv DOI: https://doi.org/10.1101/2020.02.08.926006.

  54. Cheung CY, Poon LLM, Ng IHY, Luk W, Sia SF, Wu MHS, Chan KH, Yuen Y, Gordon S, Guan Y, Peiris JSM (2005) Cytokine responses in severe acute respiratory syndrome coronavirus-infected macrophages in vitro: possible relevance to pathogenesis. J. Virol. 797819–797826. https://doi.org/10.1128/JVI.79.12.7819-7826.2005.

  55. Law et al., 2005 Law HKW, Cheung CY, Ng HY, Sia SF, Chan YO, Luk W, Nicholls JM, Peiris JSM, Lau YL (2005) Chemokine up-regulation in SARS-coronavirus-infected, monocyte-derived human dendritic cells. Blood. 106: 2366–2374. https://doi.org/ 10.1182/ blood-2004-10-4166.

  56. Chu et al., 2016 Chu H, Zhou J, Wong BHY, Li CC, Chan JFW, Cheng ZS, Yang D, Wang D, Lee ACY, Li CC, Yeung ML, Cai JP, Chan IHY, Ho WK, To KKW, Zheng BJ, Yao Y, Qin C, Yeun KY (2016) Middle east respiratory syndrome coronavirus efficiently infects human primary T lymphocytes and activates the extrinsic and intrinsic apoptosis pathways. J. Infect. Dis. 213: 904–914. https://doi.org/10.1093/infdis/jiv380.

  57. Byrnes JJ, Gross S, Ellard C, Connolly K, Donahue S, Picarella D (2009) Effects of the ACE2 inhibitor GL1001 on acute dextran sodium sulfate-induced colitis in mice. Inflamm. Res. 58: 819-827. https://doi.org/10.1007/s00011-009-0053-3.

  58. Waseem T, Duxbury M, Ito H, Ashley SW, Robinson MK (2008) Exogenous ghrelin modulates release of pro-inflammatory and anti-inflammatory cytokines in LPS-stimulated macrophages through distinct signaling pathways. Surgery 143: 334-342.

  59. Hardeland R (2018) Review Melatonin and retinoid orphan receptors: Demand for new interpretations after their exclusion as nuclear melatonin receptors. Melatonin Res.1: 78-93. DOI: 10.32794/mr11250005.

  60. Kucukakin B, Lykkesfeldt J, Nielsen HJ, Reiter RJ, Rosenberg J, Gogenur I (2008) Utility of melatonin to treat surgical stress after major vascular surgery–a safety study. J. Pineal Res. 44: 426–431. https://doi.org/10.1111/j.1600-079X.2007.00545.x.

  61. Zhao Z, Lu C, Li T, Wang W, Ye W, Zeng R, Ni L, Lai Z, Wang X, Liu C (2018) The protective effect of melatonin on brain ischemia and reperfusion in rats and humans: in vivo assessment and a randomized controlled trial. J. Pineal Res. 65: e12521, https://doi.org/10.1111/jpi.12521.

  62. Shafiei E, Bahtoei M, Raj P, Ostovar A, Iranpour D, Akbarzadeh S, Shahryari H, Anvaripour A, Tahmasebi R, Netticadan T, Movahed A (2018) Effects of N-acetyl cysteine and melatonin on early reperfusion injury in patients undergoing coronary artery bypass grafting: a randomized, open-labeled, placebo-controlled trial. Medicine 97: e11383, https://doi.org/10.1097/MD.0000000000011383.

  63. Huo X, Wang C, Yu Z, Peng Y, Wang S, Feng S, Zhang S, Tian X, Sun C, Liu K, Deng S, Ma X (2017) Human transporters, PEPT1/2, facilitate melatonin transportation into mitochondria of cancer cells: An implication of the therapeutic potential. J. Pineal Res. 62 (4): e12390. DOI: 10.1111/jpi.12390.

  64. Hevia D, González-Menéndez P, Quiros-González I, Miar A, Rodríguez-García A, Tan DX, Reiter RJ, Mayo JC, Sainz RM (2015) Melatonin uptake through glucose transporters: a new target for melatonin inhibition of cancer. J. Pineal Res. 58 (2): 234‐250. DOI:10.1111/jpi.12210.

  65. Pal PK, Maitra SK (2018) Response of gastrointestinal melatonin, antioxidants, and digestive enzymes to altered feeding conditions in carp (Catla catla). Fish Physiol. Biochem. 44: 1061–1073. https://doi.org/10.1007/s10695-018-0494-0.

  66. Reiter RJ, Calvo JR, Karbownik M, Qi W, Tan DX (2000) Melatonin and its relation to the immune system and inflammation. Ann. N. Y. Acad. Sci. 917: 376–386. https://doi.org/10.1111/j.1749-6632.2000.tb05402.x.

  67. Rodriguez C, Mayo JC, Sainz RM, Antolin I, Herrera F, Martin V, Reiter RJ (2004) Regulation of antioxidant enzymes: A significant role for melatonin. J. Pineal Res. 36: 1–9.

  68. Cuzzocrea S, Reiter RJ (2001) Pharmacological action of melatonin in shock, inflammation and ischemia/reperfusion injury. Eur. J. Pharmacol. 426: 1–10. https://doi.org/10.1016/S0014-2999(01)01175-X.

  69. Mauriz JL, Collado PS, Veneroso C, Reiter RJ, González-Gallego J (2013) A review of the molecular aspects of melatonin’s anti-inflammatory actions: recent insights and new perspectives. J. Pineal Res. 54: 1–14.doi: 10.1111/j.1600-079X.2012.01014.x.

  70. Mitra E, Bhattacharjee B, Pal PK, Ghosh AK, Mishra S, Chattopadhyay A, Bandyopadhyay D (2019) Melatonin protects against cadmium-induced oxidative damage in different tissues of rat: a mechanistic insight. Melatonin Res.2: 1-21. DOI: 10.32794/mr11250018.

  71.  Tan DX, Hardeland R (2020) Potential utility of melatonin in deadly infectious diseases related to the overreaction of innate immune response and destructive inflammation: focus on COVID-19. Melatonin Res. 3: 120-143. DOI: 10.32794/mr11250052.

  72. Ben-Nathan D, Maestroni GJ, Lustig S, Conti A (1995) Protective effects of melatonin in mice infected with encephalitis viruses. Arch. Virol. 140: 223–230.

  73. Raikhlin NT, Kvetnoy IM (1974) Lightening effect of the extract of human appendix mucosa on frog skin melanophores. Bull. Exp. Biol. Med. 8: 114–116.

  74. Fu Z, Kato H, Kotera N, Noguchi T, Sugahara K, Kubo T (2001) Regulation of hydroxyindole-O-methyltransferase gene expression in Japanese quail (Coturnixcoturnix japonica). Biosci. Biotechnol. Biochem. 65: 2504–2511. DOI: 10.1271/bbb.65.2504.

  75. Slominski A, Tobin DJ, Zmijewski MA, Wortsman J, Paus R (2008) Melatonin in the skin: synthesis, metabolism and functions. Trends Endocrinol. Metab. 19: 17–24.

  76. Hong GX, Pang SF (1995) N-Acetyltransferase activity in the quail (Coturnixcoturnix japonica) duodenum. Comp. Biochem. Physiol. 112: 251–255.

  77. Dobocovich M, Mankowsha M (2005) Functional MT1 and MT2 melatonin receptors in mammals. Endocrine 27: 101–110. DOI: 10.1385/ENDO:27:2:101.

  78. Brzozowski T, Konturek PC, Konturek SJ, Pajdo R, Bielanski W, Brzozowska I, Stachura J, Han EG (1997) The role of melatonin and L-tryptophan in prevention of acute gastric lesions induced by stress, ethanol, ischemia, and aspirin. J. Pineal Res. 23: 79-89. https://doi.org/10.1111/j.1600-079X.1997.tb00339.x.

  79. Witt-Enderby PA, Radio NM, Doctor JS, Davis VL (2006) Therapeutic treatments potentially mediated by melatonin receptors: potential clinical uses in the prevention of osteoporosis, cancer and as an adjuvant therapy. J. Pineal Res. 41: 297–305. DOI: 10.1111/j.1600-079x.2006.00369.x.

  80. Srinivasan V, Lauterbach EC, Ho KY, Acuña-Castroviejo D, Zakaria R, Brzezinski A (2012) Melatonin in antinociception: Its therapeutic applications. Curr. Neuropharmacol.  10: 167–178. DOI: 10.2174/157015912800604489.

  81. Tan DX, Reiter RJ, Manchester LC, Yan MT, El-Sawi M, Sainz RM, Mayo JC, Kohen R, Allegra M, Hardeland R (2002) Chemical and physical properties and potential mechanisms: melatonin as a broad spectrum antioxidant and free radical scavenger. Curr. Top. Med. Chem. 2: 181-97. https://www.ncbi.nlm.nih.gov/pubmed /11899100.

  82. Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, Qin L (2016) Melatonin as an antioxidant: under promises but over delivers. J. Pineal Res. 61: 253–278. DOI: 10.1111/jpi.12360.

  83. Tan DX, Reiter RJ (2019) Mitochondria: the birth place, battle ground and the site of melatonin metabolism in cells. Melatonin Res. 2: 44-66. DOI: https://doi.org/10.32794/ mr11250011.

  84. Reiter RJ, Tan DX, Manchester LC, Qi W. (2001) Biochemical reactivity of melatonin with reactive oxygen and reactive nitrogen species: A review of the evidence. Cell Biochem. Biophys. 34: 237–256. DOI: 10.1385/CBB:34:2:237.

  85. Cuzzocrea S, Reiter JR (2002) Pharmacological action of melatonin in acute and chronic inflammation. Curr. Top. Med. Chem. 2: 153–165.

  86. Nosál'ová et al., 2007 Nosál'ová V, Zeman M, Černá S, Navarová J, Zakálová M (2007) Protective effect of melatonin in acetic acid induced colitis in rats. J. Pineal Res. 42: 364–370. DOI: 10.1111/j.1600-079X.2007.00428.x.

  87. Torres-Farfan C, Richter HG, Rojas-García P, Vergara M, Forcelledo ML, Valladares LE, Torrealba F, Valenzuela GJ, Seron- Ferre M. (2003) mt1 Melatonin receptor in the primate adrenal gland: inhibition of adrenocorticotropin-stimulated cortisol production by melatonin. J. Clin. Endocrinol. Metab. 88: 450–458. DOI: 10.1210/jc.2002-021048.

  88. Saito S, Tachibana T, Choi YH, Denbow DM, Furuse M (2005) ICV melatonin reduces stress responses in neonatal chicks. Behav. Brain Res. 165: 197–203. DOI: 10.1016/j.bbr.2005.06.045.

  89. Lopez-Burillo S, Tan DX, Rodriguez-Gallego V, Manchester LC, Mayo JC, Sainz RM, Reiter RJ (2003) Melatonin and its derivatives cyclic 3-hydroxymelatonin, N1-acetyl-N2-formyl-5- methoxykynuramine, and 6-hydroxymelatonin reduced oxidative damage induced by Fenton reagents. J. Pineal Res. 34: 237–256. DOI: 10.1034/j.1600-079x.2003.00025.x.

  90. Imai Y, Kuba K, Neely G, Yaghubian-Malhami R, Perkmann T, van Loo G, Ermolaeva M, Veldhuizen R, Leung YHC, Wang H, Liu H, Sun Y, Pasparakis M, Kopf M, Mech C, Bavari S, Peiris JSM, Slutsky AS, Akira S, Hultqvist M, Holmdahl R, Nicholls J, Jiang C, Binder CJ, Penninger JM (2008) Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell 133: 235–249, https://doi.org/10.1016/j.cell.2008.02.043.

  91. Mosca A, Leclerc M, Hugot JP (2016) Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem? Front. Microbiol. 7: 1–12. https://doi.org/10.3389/fmicb.2016.00455.

  92. Khan I, Ullah N, Zha L, Bai Y, Khan A, Zhao T, Che T, Zhang C (2019) Alteration of gut microbiota in inflammatory bowel disease (IBD): cause or consequence? IBD treatment targeting the gut microbiome. Pathogens (3): 1–28. https://doi.org/10.3390/ pathogens 8030126.

  93. Bingula R, Filaire M, Radosevic-Robin N, Bey M, Berthon JY, Bernalier-DonadilleA, Vasson MP, Filaire E (2017) Desired turbulence? Gut-lung Axis, immunity, and lung Cancer. J. Oncol. 2017: 5035371. https://doi.org/10.1155/2017/5035371.

  94. Zhang D, Li S, Wang N, Tan HY, Zhang Z, Feng Y (2020) The cross-talk between gut microbiota and lungs in common lung diseases. Front. Microbiol. 11: 1–14. https://doi.org/10.3389/fmicb. 2020.00301.

  95. Keely S, Talley NJ, Hansbro PM (2012) Pulmonary-intestinal cross-talk in mucosal inflammatory disease. Mucosal Immunol. (1): 7–18. https://doi.org/10.1038/mi.2011.55.

  96. Dumas A, Bernard L, Poquet Y, Lugo-Villarino G, Neyrolles O (2018) The role of the lung microbiota and the gut–lung axis in respiratory infectious diseases. Cell. Microbiol.20(12): e12966. https://doi.org/10.1111/cmi.12966.

  97. Dickson RP, Arbor A (2017) The microbiome and critical illness. Lancet Respir. Med. (1): 59–72. https://doi.org/10.1016/S2213-2600(15)00427-0.

  98. Negi S, Das DK, Pahari S, Nadeem S, Agrewala JN (2019) Potential role of gut microbiota in induction and regulation of innate immune memory. Front. Immunol10: 1–12. https://doi.org/10.3389/fimmu.2019.02441.

  99. Round JL, Mazmanian SK (2010) Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. 107 (27): 12204–12209. https://doi.org/10.1073/pnas.0909122107.

  100. Domínguez-Díaz C, García-Orozco A, Riera-Leal A, Padilla-Arellano JR, Fafutis-Morris M (2019) Microbiota and its role on viral evasion: Is it with us or against us? Front. Cell. Infect. Microbiol. 9: 256. https://doi.org/10.3389/fcimb.2019.00256.

  101. Dhara D, Mohanty A (2020) Gut microbiota and Covid-19- possible link and implications. Virus Res. 285: 198018. https://doi.org/10.1016/j.virusres.2020.198018.

  102. Groves HT, Higham SL, Moffatt MF, Cox MJ, Tregoning JS (2020) Respiratory viral infection alters the gut microbiota by inducing inappetence. mBio. 11: e03236-19. https://doi.org/10.1128/mBio.03236-19.

  103. Zuo T, Zhang F, Lin GCY, Yeoh YK, Li AYL, Zhan H, Wan Y, Chung A, Cheung CP, Chen N, Lai CKC, Chen Z, Fung ESC, Chan V, Ling L, Joynt G, Hui DSC, Ng AC (2020) Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology https://doi.org/10.1053/j.gastro.2020.05.048.

  104. Hajifathalian K, Mahadev S, Schwartz RE, Shah S, Sampath K, Schnoll-Sussman F, Brown Jr RS, Carr-Locke D, Cohen DE, Sharaiha RZ (2020) SARS-COV-2 infection (coronavirus disease 2019) for the gastrointestinal consultant. World J. Gastroenterol. 26 (14): 1546–1553. DOI: 10.3748/wjg.v26.i14.1546.

  105. Lin L, Jiang X, Zhang Z, Huang S, Zhang Z, Fang Z, Gu Z, Gao L, Shi H, Mai L, Liu Y, Lin X, Lai R, Yan Z, Li X, Shan H (2020) Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut69 (6): 997‐1001. DOI: 10.1136/gutjnl-2020-321013.

  106. Paulose JK, Cassone VM (2016) The melatonin-sensitive circadian clock of the enteric bacterium Enterobacter aerogenesGut Microbes. 7: 424-427. DOI: 10.1080/19490976. 2016.1208892.

  107. Yin J, Li Y, Han H, Chen S, Gao J, Liu G, Wu X, Deng J, Yu Q, Huang X, Fang R (2018) Melatonin reprogramming of gut microbiota improves lipid dysmetabolism in high fat diet fed mice. J. Pineal Res. 65: e12524. DOI: 10.1111/jpi.12524.

  108. Zhu D, Ma Y, Ding S, Jiang H, Fang J (2018) Effects of melatonin on intestinal microbiota and oxidative stress in colitis mice. Biomed. Res. 2018: 2607679. DOI: 10.1155/2018/2607679.

  109. Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, Li T and Yin Y (2018) Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front. Cell Infect. Microbiol. 8: 13. DOI: 10.3389/fcimb.2018.00013.

  110. Chojnacki C, Wiśniewska-Jarosińska M, Kulig G, Majsterek I, Reiter RJ, Chojnacki J (2013) Evaluation of enterochromaffin cells and melatonin secretion exponents in ulcerative colitis. World J. Gastroenterol. 19: 3602-3607. DOI: 10.3748/wjg.v19. i23.3602.

  111. Galano A, Tan DX, Reiter RJ (2011) Melatonin as a natural ally against oxidative stress: a physicochemical examination. J. Pineal Res. 51: 1–16. DOI: 10.1111/j.1600-079X.2011.00916.x.

  112. Carrillo-Vico A, Lardone PJ, Alvarez-Sánchez N, Rodríguez-Rodríguez A, Guerrero JM (2013) Melatonin: buffering the immune system. Int. J. Mol. Sci.14: 8638–8683. DOI: 10.3390/ijms14048638.

  113. Dong Y, Fan C, Hu W, Jiang S, Ma Z, Yan X, Deng C, Di S, Xin Z, Wu G, Yang Y, Reiter RJ, Liang G (2016) Melatonin attenuated early brain injury induced by subarachnoid hemorrhage via regulating NLRP3 inflammasome and apoptosis signalling. J. Pineal Res. 60: 253–262. DOI: 10.1111/jpi.12300.

  114. Reiter RJ, Tan DX, Acuna-Castroviejo D, Burkhardt S, Karbownik M (2000) Melatonin: Mechanisms and actions as an antioxidant. Curr. Top. Biophys. 24: 171–183.

  115. Vriend J, Reiter RJ (2014) Melatonin as a proteasome inhibitor. Is there any clinical evidence? Life Sci.115: 8–14. DOI: 10.1016/j.lfs.2014.08.024.

  116. Hu ZP, Fang XL, Fang N, Wang XB, Qian HY, Cao Z, Cheng Y, Wang BN, Wang Y (2013) Melatonin ameliorates vascular endothelial dysfunction, inflammation, and atherosclerosis by suppressing the TLR4/NF-kB system in high-fat-fed rabbits. J. Pineal Res. 55: 388–398. DOI: 10.1111/jpi.12085.

  117. Najafi M, Shirazi A, Motevaseli E, Rezaeyan AH, Salajegheh A, Rezapoor S (2017) Melatonin as an anti-inflammatory agent in radiotherapy. Inflammopharmacol. 25: 403-413. DOI 10.1007/s10787-017-0332-5.

  118. Akinrinmade FJ, Akinrinde AS, Amid A (2016) Changes in serum cytokine levels, hepatic and intestinal morphology in aflatoxin B1-induced injury: modulatory roles of melatonin and flavonoid-rich fractions from ChromolenaodorataMycotoxin Res. 32: 53-60.DOI 10.1007/s12550-016-0239-9.

  119. Carrillo-Vico A, García-Mauriño S, Calvo JR, Guerrero JM (2003) Melatonin counteracts the inhibitory effect of PGE2 on IL-2 production in human lymphocytes via its mt1 membrane receptor. FASEB J. 17: 755–757.

  120. Maldonado MD, García-Moreno H, González-Yanes C, Calvo JR (2016) Possible involvement of the inhibition of NF-κB factor in anti-inflammatory actions that melatonin exerts on mast cells.  J. Cell. Biochem.1 17 (8):1926-33. DOI: 10.1002/jcb.25491.

  121. Reiter RJ (2000) Melatonin: Lowering the high price of free radicals. News Physiol. Sci. 15: 246–250.

  122. Hardeland R, Cardinali DP, Srinivasan V, SpenceDW, Brown GM, Pandi-Perumal SR, (2011) Melatonin—a pleiotropic, orchestrating regulator molecule. Progress Neurobiol. 93: 350–384.

  123. Favero G, Franceschetti L, Bonomini F, Rodella LF, Rezzani R (2017) Melatonin as an anti-inflammatory agent modulating inflammasome activation. Int. J. Endocrinol. 2017: 1835195. https://doi.org/10.1155/2017/1835195.

  124. Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, Xie C, Ma K, Shang K, Wang W, Tian D (2020) Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. DOI: 10.1093/cid/ciaa248.

  125. Neurath MF (2020) COVID-19 and immunomodulation in IBD. Gut. 69: 1335–1342. DOI: 10.1136/gutjnl-2020-321269.

  126. Santaolalla R, Fukata M, Abreu MT (2011) Innate immunity in the small intestine. Curr. Opin. Gastroenterol.27 (2): 125–131. DOI:10.1097/MOG.0b013e3283438dea.

  127. Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, Wang Z, Li J, Li J, Feng C, Zhang Z, Wang L, Peng L, Chen L, Qin Y, Zhao D, Tan S, Yin L, Xu J, Zhou C, Jiang C, Liu L (2020) Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci. China Life Sci. https://doi.org/10.1007/s11427-020-1643-8.

  128. Xiao F, Tang M, Zheng X,  Liu Y, Li X, Shan H (2020) Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology. 158 (6): 1831–1833. DOI: https://doi.org/10.1053/j.gastro.2020.02.055.

  129. Girardin SE, Boneca IG, Carneiro LA, Antignac A, Jehanno M, Viala J, Tedin K, Taha MK, Labigne A, Zahringer U, Coyle AJ, DiStefano PS, Bertin J, Sansonetti PJ, Philpott DJ (2003) Nod1 detects a unique muropeptide from gram-negative bacterial peptidoglycan. Science 300: 1584-1587. DOI: 10.1126/science.1084677.

  130. Kim JG, Lee SJ, Kagnoff MF (2004) Nod1 is an essential signal transducer in intestinal epithelial cells infected with bacteria that avoid recognition by Toll-like receptors. Infect. Immun. 72: 1487–1495. DOI: 10.1128/IAI.72.3.1487-1495.2004.

  131. Pedra JH, Cassel SL, Sutterwala FS (2009) Sensing pathogens and danger signals by the inflammasome. Curr. Opin. Immunol. 21: 10–16. DOI: 10.1016/j.coi.2009.01.006.

  132. Stutz A, Golenbock DT, Latz E (2009) Inflammasomes: too big to miss. J. Clin. Invest. 119: 3502–3511. DOI: 10.1172/JCI40599.

  133. Zhang R, Wanga X, Nia L, Dia X, Maa B, Niu S, Liua C, Reiter RJ (2020) COVID-19: Melatonin as a potential adjuvant treatment. Life Sci. 250: 117583. https://doi.org/10.1016/j.lfs.2020.117583.

  134. Gu J, Korteweg C (2007) Pathology and pathogenesis of severe acute respiratory syndrome. Am. J. Pathol. 170 (4): 1136–1147. DOI: 10.2353/ajpath.2007.061088.

  135. Gu J, Gong EC, Zhang B, Zheng J, Gao ZF, Zhong YF, Zou WZ, Zhan J, Wang SL, Xie ZG, Zhuang H, Wu BQ, Zhong HH, Shao HQ, Fang WG, Gao DX, Pei F, Li XW, He ZP, Xu DZ, Shi XY, Anderson VM, Leong ASY (2005) Multiple organ infection and the pathogenesis of SARS. J. Exp. Med. 202: 415-424. DOI: 10.1084/jem.20050828.

  136. Shi X, Gong E, Gao D, Zhang B, Zheng J, Gao Z, Zhong Y, Zou W, Wu B, Fang W, Liao S, Wang S, Xie Z, Lu M, Hou L, Zhong H, Shao H, Li N, Liu C, Pei F, Yang J, Wang Y, Han Z, Shi X, Zhang Q, You J, Zhu X, Gu J (2005) Severe acute respiratory syndrome associated coronavirus is detected in intestinal tissues of fatal cases. Am. J. Gastroenterol. 100: 169-176. DOI: 10.1111/j.1572-0241.2005.40377.x.

  137. Leung WK, To KF, Chan PK, Chan HL, Wu AK, Lee N, Yuen KY, Sung JJ (2003) Enteric involvement of severe acute respiratory syndrome-associated coronavirus infection. Gastroenterology 125: 1011–1017.DOI: 10.1016/s0016-5085(03)01215-0.

  138. He L, Ding Y, Zhang Q, Che X, He Y, Shen H, Wang H, Li Z, Zhao L, Geng J, Deng Y, Yang L, Li J, Cai J, Qiu L, Wen K, Xu X, Jiang S (2006) Expression of elevated levels of pro-inflammatory cytokines in SARSCoV- infected ACE2(+) cells in SARS patients: relation to the acute lung injury and pathogenesis of SARS. J. Pathol. 210: 288–297. DOI: 10.1002/path.2067.

  139. Miller SC, Pandi-Perumal SR, Esquifino AI, Cardinali DP, Maestroni GJM (2006) The role of melatonin in immuno-enhancement: potential application in cancer. Int. J. Exp. Pathol. 87: 81–87. https://doi.org/10.1111/j.0959-9673.2006.00474.x.

  140. Kaur C, Ling EA (1999) Effects of melatonin on macrophages/microglia in postnatal rat brain. J. Pineal Res. 26: 158–168. https://doi.org/10.1111/j.1600-079x.1999.tb00578.x.

  141. Park et al., 2015 Park YS, Chung SH, Lee SK, Kim JH, Kim JB, Kim TK, Kim DS, Baik HW (2015) Melatonin improves experimental colitis with sleep deprivation. Int. J. Mol. Med. 35: 979-986. DOI: 10.3892/ijmm.2015.2080.

  142. Sanchez-Lopez AL, Ortiz GG, Pacheco-Moises FP, Mireles-Ramirez MA, Bitzer-Quintero OK, Delgado-Lara DLC, Ramirez-Jirano LJ, Velazquez- Brizuela IE (2018) Efficacy of melatonin on serum pro-inflammatory cytokines and oxidative stress markers in relapsing remitting multiple sclerosis. Arch. Med. Res. 49: 391–398. https://doi.org/10.1016/j.arcmed.2018.12.004.

  143. Bazyar H, Gholinezhad H, Moradi L, Salehi P, Abadi F, Ravanbakhsh M, Zare Javid A (2019) The effects of melatonin supplementation in adjunct with non-surgical periodontal therapy on periodontal status, serum melatonin and inflammatory markers in type 2 diabetes mellitus patients with chronic periodontitis: a double-blind, placebo-controlled trial. Inflammopharmacol. 27: 67–76. https://doi.org/10.1007/s10787-018-0539-0.

  144. Tahan et al., 2011 Tahan G, Gramignoli R, Marongiu F, Aktolga S, Cetinkaya A, Tahan V, Dorko K (2011) Melatonin expresses powerful anti-inflammatory and antioxidant activities resulting in complete improvement of acetic-acid-induced colitis in rats. Dig. Dis. Sci. 56: 715-720. DOI: 10.1007/s10620-010-1364-5.

  145. Volt H, García JA, Doerrier C, Díaz-Casado ME, Guerra-Librero A, López LC, Escames G, Tresguerres JA, Acuña-Castroviejo D (2016) Same molecule but different expression: aging and sepsis trigger NLRP3 inflammasome activation, a target of melatonin. J. Pineal Res. 60: 193–205. DOI: 10.1111/jpi.12303.

  146. Zarezadeh M, Khorshidi M, Emami M, Janmohammadi P, Kord-Varkaneh H, Mousavi SM, Mohammed SH, Saedisomeolia A, Alizadeh S (2019) Melatonin supplementation and pro-inflammatory mediators: a systematic review and meta-analysis of clinical trials, Eur. J. Nutr. https://doi.org/10.1007/s00394-019-02123-0.

  147. Bourne RS, Mills GH, Minelli C (2008) Melatonin therapy to improve nocturnal sleep in critically ill patients: encouraging results from a small randomised controlled trial, Crit. Care12: R52. https://doi.org/10.1186/cc6871.

  148.  Mistraletti G, Umbrello M, Sabbatini G, Miori S, Taverna M, Cerri B, Mantovani ES, Formenti P, SpanuP, D’Agostino A, Salini S, Morabito A, Fraschini F, Reiter RJ, Iapichino G (2015) Melatonin reduces the need for sedation in ICU patients: a randomized controlled trial. Minerva Anestesiol. 81: 1298–1310.

  149. Andersen LPH, Gogenur I, Rosenberg J, Reiter RJ (2016) The safety of melatonin in humans. Clin. Drug Investig. 36: 169–175. https://doi.org/10.1007/s40261-015-0368-5.

  150. Nordlund JJ, Lerner AB (1977) The effects of oral melatonin on skin color and on the release of pituitary hormones. J. Clin. Endocrinol. Metab. 45: 768–774. https://doi.org/10.1210/jcem-45-4-768.

  151. Sun CK, Lee FY, Kao YH, Chiang HJ, Sung PH, Tsai TH, Lin YC, Leu S, Wu YC, Lu HI, Chen YL, Chung SY, Su HL, Yip HK (2015) Systemic combined melatonin-mitochondria treatment improves acute respiratory distress syndrome in the rat. J. Pineal Res. 58: 137–150. https://doi.org/10.1111/jpi.12199.

  152. Reiter RJ, Abreu-Gonzalez P, Marik PE, Dominguez-Rodriguez A (2020) Therapeutic algorithm for use of melatonin in patients with COVID-19. Front Med. 7: 226. DOI: 10.3389/fmed.2020.00226.

  153. Giménez VMM, Inserra F, Tajer CD, Mariani J, Ferder L, Reiter RJ, Manucha W (2020) Lungs as target of COVID-19 infection: Protective common molecular mechanisms of vitamin D and melatonin as a new potential synergistic treatment. Life Sci254: 117808. DOI: 10.1016/j.lfs.2020.117808.


 CCBY-1.png

This work is licensed under a Creative Commons Attribution 4.0 International License