Begüm Öğünç, Nur Ramoğlu, Irmak Sakin

Acıbadem University School of Medicine, Istanbul, Turkey

Keywords: Alzheimer’s disease, inflammatory bowel disease, methylene blue

Abstract

High-efficiency oxidative phosphorylation plays a key role in the progression of many diseases. There is growing evidence of methylene blue’s protective and reversing actions against neurodegenerative and inflammatory bowel diseases through different mechanisms. In Alzheimer’s disease (AD), oligomeric amyloid beta accumulates in the mitochondria and contributes to mitochondrial dysfunction, which occurs before significant plaque deposition. Methylene blue provides an alternative mitochondrial electron transfer pathway, switching from high-efficiency oxidative phosphorylation to the low-efficiency aerobic glycolysis pathway by receiving electrons from NADH in the presence of complex I and transferring them to cytochrome C. The second mechanism is the inhibition of active caspases, especially Caspase-6, a cysteinyl protease causing inflammation and cell death, which has been associated with age-dependent cognitive decline and the pathology of sporadic and familial AD. The third mechanism is the reversal of tau aggregation by oxidizing cysteine residues in tau and forming a more stable monomer, thus blocking tau-tau bindings as well as clearing tau pathology through increased autophagy. In regards to inflammatory bowel disease, reducing oxidative stress and attenuating inflammatory pathways inhibits epithelial destruction in acetic acid-induced colitis. Methylene blue has an anti-colitis effect, mainly relying on its mitochondrial efficacy-restoring, antioxidative, anti-inflammatory, and anti-apoptotic properties. In summary, methylene blue is a promising agent for both AD and inflammatory bowel disease due to its beneficial effects as well as its low cost and high accessibility.

Methylene blue (MB), a heterocyclic phenothiazine-based aromatic chemical compound, was originally synthesized as a textile dye but gained a significant role in the treatment of various bacterial and viral infections, methemoglobinemia, carbon monoxide poisoning, vasoplegic syndrome, and ifosfamide neurotoxicity, as well as in the surgical staining of the parathyroid glands or fistula. The repurposing of previously established drugs is being substantially considered in modern pharmacology, in terms of time- and money-saving features in addition to their previously achieved extensive safety testing records. Consequently, MB has become a promising drug in the treatment and decelerating the progression of certain diseases, such as Alzheimer’s disease (AD) and inflammatory bowel disease (IBD).[1]

PubMed, Google Scholar, and UpToDate were searched for the keywords “methylene blue”, “Alzheimer’s disease”, “inflammatory bowel diseases”, “methylene blue and Alzheimer’s disease”, and “methylene blue and inflammatory bowel disease”. Abstracts were read by the authors of the present article and the most relevant studies and reviews were analyzed. After the overlapping studies and reviews were excluded, the remaining 62 articles were grouped according to their topics and the similarity of their contents.

METHYLENE BLUE AND ALZHEIMER’S DISEASE

Alzheimer's disease is a progressive neurodegenerative disorder causing severe disability or death and reduces the quality of life for both patients and families. Alzheimer's disease remains the most common cause of dementia worldwide, and its prevalence continues to rise due to the aging world population, affecting 1 in 9 people over the age of 65 in the United States. This rate approximately matches the estimated prevalence in Turkey.[1-3] As it stands, there is no effective treatment for AD that can halt or significantly slow the patient’s inevitable decline. In this regard, this makes AD unique among the world’s major killers, as it is one of the few top causes of death with little to no preventative or therapeutic measures.[1]

Alzheimer's disease is histologically characterized by the classical accumulation of senile plaques and neurofibrillary tangles (NFT), consisting of hyperphosphorylated tau protein[4] and amyloid beta (Aβ) oligomers, which are products of the proteolytic cleavage of membrane-bound amyloid precursor protein (APP) by &gamma-secretase and β-secretase. Amyloid precursor protein processing of Aβ can proceed down one of two pathways, the first of which is the non-amyloidogenic pathway, which starts with APP cleavage by α-secretase that yields a soluble N-terminal fragment, sAPP-α, and a membrane-bound C-terminal fragment (CTF-α). CTF-α is further processed by γ-secretase to yield another N-terminal soluble fragment, p3. The soluble fragments generated by this sequence do not form aggregates and are not neurotoxic. However, the second pathway, the amyloidogenic pathway, commences with APP cleavage by β-secretase, generating sAPP-β. γ-secretase cleavage of the remaining CTF-β fragment generates Aβ, which progressively aggregates from oligomers to plaques that accumulate extracellularly, that interfere with cellular function and activate inflammatory pathways.[5] Oligomeric Aβ also accumulates intracellularly, localizing in the mitochondria, and contributes to mitochondrial dysfunction and energy deficiency in AD pathology.[6]

Until now, the main approach to AD therapeutic research targeted Aβ. As recently as 2007, MB has begun to be studied in this capacity.[7,8] According to the studies investigating its treatment capacity, MB was found to be promising due to certain actions, one of them being improving cognitive function, mainly by promoting complex IV activity and mitochondrial activity.[9] Mitochondrial dysfunction is found to be present before significant plaque deposition and cognitive decline,[10-12] which makes MB a promising agent in the early treatment of AD through its complex IV activity via its electron cycling mechanism as well as its upregulating effect on heme synthesis.[9,13]

The mitochondrial activity of MB has been further investigated in another study, resulting in the discovery of its ability to provide an alternative electron transfer pathway. In this pathway, MB receives electrons from NADH in the presence of complex I and delivers them to cytochrome C. A switch from high-efficiency oxidative phosphorylation to the low-efficiency aerobic glycolysis pathway (Warburg effect) occurs, increasing oxygen consumption, decreasing glycolysis, and increasing in vitro glucose uptake, as well as enhancing glucose uptake and regional cerebral blood flow in rats upon acute treatment. In summary, there is growing evidence that enhancement of mitochondrial oxidative phosphorylation via alternative mitochondrial electron transfer may offer protective action against neurodegenerative diseases.[14]

Several studies have demonstrated that MB improves cognitive deficits induced by hippocampal damage by upregulating complex IV and ATP production, decreasing oxidative stress markers through methods such as electron cycling, and inhibiting downstream mechanisms and interactions such as the inhibition of Aβ-ABAD binding, therefore preventing the production of associated reactive oxygen species (ROS), matrix metalloproteinase (MMP) failure, and subsequent cell death,[15-18] as well as the cytosolic release of pro-death factors in several AD models.[15,19,20]

Another study investigated the tau cysteine oxidation mechanism of MB to clarify whether MB may modulate the activity of caspases. Caspases are a group of cysteinyl proteases involved in inflammation and cell death. One of the effector caspases, Caspase-6 (Casp6), was found to be strongly associated with agedependent cognitive decline and the underlying pathology of sporadic and familial AD.[21-28] Results of the study showed that MB and its derivatives efficiently inhibited active caspases in vitro, in cells, and in vivo at concentrations allowing phenothiazine-mediated Tau disaggregation. These results indicated that MB, or its derivatives,[21,29] may have additional effects in AD by inhibiting caspases,[21,30] preventing caspase activation in other degenerative conditions,[21,31] and predisposing individuals undergoing chronic treatment to cancer via the inhibition of effector caspase-3.

Another key factor in AD pathophysiology is the presence of neurofibrillary tangles (NFT), which are aggregates of hyperphosphorylated tau protein (p-tau). Tau is a microtubule-associated protein that stabilizes neuronal microtubules as a natural and prevalent component of CNS. Tau is phosphorylated by kinases such as glycogen-synthase kinase 3 β (GSK3β), c-Jun kinase (JNK), and cyclin-dependent kinase 5 (cdk5)[32-34] as a normal component of the physiological process. However, this process dissociates tau from microtubules and destabilizes them, disrupting cellular function and axonal transport as well as mitochondrial transport as mentioned above. Nevertheless, the excessive phosphorylation of tau aggregates and forms tangles, as the amount of p-tau exceeds the proteasome’s clearance capacity. However, it is still under debate whether taupathy precedes Aβ pathology or is a consequence.[35-37]

Targeting this pathological finding, MB was found to reverse tau aggregation by blocking tau-tau binding as a consequence of inhibiting filament formation on the first and fourth repeat peptides on the tau microtubule binding domain[31,38,39] and prevent fibrillization by oxidizing cysteine residues in tau resulting in the formation of a more stable monomer form that is resistant to aggregation.[23] Another study showed that MB clears the tau pathology through increased autophagy in JNPL3 organotypic slices, by interacting with and promoting the oxidation of tau cysteine residues, thus preventing the formation of fibrils and their toxic precursors.[21,23]

Lastly, MB was found to inhibit the aggregation of proteins that take on β-sheet formation in vitro, including tau.[31,39] Methylene blue trials in vivo have yielded mixed results. In a mouse tauopathy model, 1 mM MB was administered to the right hippocampus using a mini-osmotic pump. This resulted in a decrease in total tau levels paralleled by a decrease in phosphorylated (S202/T205) tau, as well as an improvement in behavior but without any remarkable effect on pathology.[41] When the mice were administered a therapeutically relevant dose of 10 mg/kg of MB in their drinking water for 12 weeks, there was a variable improvement in behavior and a decrease in soluble tau levels, with no change in tau tangle pathology. Interestingly, when the levels of MB in the cerebellum were measured, they were found to positively correlate with Morris water maze performance and inversely correlate with soluble tau levels. These results were compatible with previous findings suggesting that NFTs are likely not associated with functional deficits but that reducing soluble tau levels is beneficial.[40] These results also suggested that the putative therapeutic effect of MB may not be associated with its ability to inhibit aggregate formation. In zebrafish, MB inhibited the aggregation of a mutant form of huntingtin gene, but had no effect on tau pathology when an FTDP-17 mutant tau was expressed, similar to what was observed in the mouse model.[41]

However, in contrast to the mouse model, MB treatment in zebrafish did not prevent or reverse the functional deficits that resulted from the expression of the FTDP-17 mutant tau or the mutant huntingtin.[41]

According to another study that reported a correlation between MB and taupathy, MB was found to clear tau filament by inducing autophagy[42] by reducing tau load in different taupathy transgenic mouse models over shortterm and long-term treatment, and this clearance was associated with improvement of cognitive deficits.[43,44] A more stable variant of the MB in reduced form, Leuco-Methylthioninium Bis (Hydromethanesulphonate) (LMTM), was developed[45] for use in human clinical trials to show both cognitive and cerebral blood flow improvements[46] under the name “REMBER”. Unfortunately, the study yielded negative results.[47]

METHYLENE BLUE AND INFLAMMATORY BOWEL DISEASE

Inflammatory bowel disease is a term mainly used to describe two conditions: ulcerative colitis (UC) and Crohn's disease (CD). Ulcerative colitis and CD are chronic conditions that involve inflammation of the gut. Ulcerative colitis only affects the colon (large intestine), while Crohn's disease can affect any part of the digestive system, from the mouth to the anus.[48] It is estimated that over 1 million residents in the USA and 2.5 million in Europe have IBD, with substantial expenses spent on health care.[49]

Although IBD has been thought to be idiopathic, it has two main attributable causes, including genetic and environmental factors. The gastrointestinal tract where this disease occurs is central to the immune system, and the innate and the adaptive immune systems are balanced in complex interactions with intestinal microbes under homeostatic conditions. However, in IBD, this homeostasis is disrupted and uncontrolled intestinal inflammation occurs.[50]

Methylene blue is conventionally used for staining in endocytoscopy (EC), one of the most novel endoscopic diagnostic procedures that provides up to 1,150 times optical magnification of the gastrointestinal mucosa. This approach allows real-time visualization of tissue and cellular structure. Endocytoscopy, along with confocal laser endomicroscopy, is considered in vivo “optical biopsy”[51] as it enables the visualization of cellular atypia of gastrointestinal mucosae[52] and detection of dysplasia or neoplasia.[53] A study on UC patients reported that chromoendoscopyguided endomicroscopy decreases the number of biopsies performed and significantly increases the per-biopsy yield of intraepithelial neoplasia. Endomicroscopy is an accurate tool for intraepithelial neoplasia detection.[54]

Several pro-inflammatory cytokines are involved in the progression of IBD. One of them, IL-6, activates signal transducer and activator of transcription 3 (STAT3) and has an important function in the inflammatory response. There is elevated production of IL-6 and its soluble IL-6 receptor in UC and CD patients.[55,56] Interleukin-6 also has a key role in the pathogenesis of UC and the carcinogenesis of colorectal cancers related to UC.[57] Moreover, it has been reported that IL-17 can increase the recruitment of T cells into the lamina propria during the inflammatory response.

A previous study revealed that MB can efficiently inhibit epithelial destruction in acetic acid-induced colitis by decreasing oxidative stress and attenuating inflammatory pathways.[58] Other previous studies observed decreased expression of Bcl-2 and elevated levels of Bax protein in UC. These changes were significantly reversed by MB, demonstrating its anti-apoptotic effects. The overproduction of reactive oxygen species in neuronal cells can result in the collapse of cellular components, especially DNA, leading to cell death.[59] This indicates that the anti-apoptotic properties of MB may be partially due to its antioxidative effect.

In a study by El Sayed and Sayed,[60] the anti-colitis effect of MB was tested in a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis model. According to the study, the role of mitochondrial dysfunction is significant in the pathogenesis of TNBS-induced UC. On the other hand, MB restores mitochondrial efficacy and exerts antioxidative, anti-inflammatory, and anti-apoptotic effects. These results indicate that MB is a promising therapeutic agent for the management of UC without harmful side effects.[60] As there are overlaps in the pathogenesis of UC and CD, the mechanisms suggest that MB could also be administered in CD treatment and further related research is warranted.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Tucker D, Lu Y, Zhang Q. From Mitochondrial Function to Neuroprotection-an Emerging Role for Methylene Blue. Mol Neurobiol 2018;55:5137-53.
  2. Weller J, Budson A. Current understanding of Alzheimer's disease diagnosis and treatment. F1000Res 2018;7:F1000 Faculty Rev-1161.
  3. Saka Topçuoğlu E, Selekler K. Alzheimer hastalığı. Geriatri 1998;1:63-7.
  4. Zhao M, Liang F, Xu H, Yan W, Zhang J. Methylene blue exerts a neuroprotective effect against traumatic brain injury by promoting autophagy and inhibiting microglial activation. Mol Med Rep 2016;13:13-20.
  5. Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci 2007;120:4081-91.
  6. Arrázola MS, Silva-Alvarez C, Inestrosa NC. How the Wnt signaling pathway protects from neurodegeneration: the mitochondrial scenario. Front Cell Neurosci 2015;9:166.
  7. Irwin JA, Wong HE, Kwon I. Different fates of Alzheimer's disease amyloid-b fibrils remodeled by biocompatible small molecules. Biomacromolecules 2013;14:264-74.
  8. Necula M, Breydo L, Milton S, Kayed R, van der Veer WE, Tone P, et al. Methylene blue inhibits amyloid Abeta oligomerization by promoting fibrillization. Biochemistry 2007;46:8850-60.
  9. Atamna H. Amino acids variations in amyloidbeta peptides, mitochondrial dysfunction, and new therapies for Alzheimer's disease. J Bioenerg Biomembr 2009;41:457-64.
  10. Supnet C, Bezprozvanny I. Neuronal calcium signaling, mitochondrial dysfunction, and Alzheimer's disease. J Alzheimers Dis 2010;20 Suppl 2(Suppl 2):S487-98.
  11. Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD. Mitochondrial bioenergetic deficit precedes Alzheimer's pathology in female mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2009;106:14670-5.
  12. Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, et al. Mitochondrial abnormalities in Alzheimer's disease. J Neurosci 2001;21:3017-23.
  13. Atamna H, Kumar R. Protective role of methylene blue in Alzheimer's disease via mitochondria and cytochrome c oxidase. J Alzheimers Dis 2010;20 Suppl 2:S439-52.
  14. Yang SH, Li W, Sumien N, Forster M, Simpkins JW, Liu R. Alternative mitochondrial electron transfer for the treatment of neurodegenerative diseases and cancers: Methylene blue connects the dots. Prog Neurobiol 2017;157:273-91.
  15. Zakaria A, Hamdi N, Abdel-Kader RM. Methylene blue improves brain mitochondrial ABAD functions and decreases Ab in a neuroinflammatory Alzheimer’s disease mouse model. Mol Neurobiol 2016;53:1220-8.
  16. Lim YA, Grimm A, Giese M, Mensah-Nyagan AG, Villafranca JE, Ittner LM, et al. Inhibition of the mitochondrial enzyme ABAD restores the amyloidb-mediated deregulation of estradiol. PLoS One 2011;6:e28887.
  17. Takuma K, Yao J, Huang J, Xu H, Chen X, Luddy J, et al. ABAD enhances Abeta-induced cell stress via mitochondrial dysfunction. FASEB J 2005;19:597-8.
  18. Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, Wang N, et al. ABAD directly links Abeta to mitochondrial toxicity in Alzheimer's disease. Science 2004;304:448-52.
  19. Violet M, Chauderlier A, Delattre L, Tardivel M, Chouala MS, Sultan A, et al. Prefibrillar Tau oligomers alter the nucleic acid protective function of Tau in hippocampal neurons in vivo. Neurobiol Dis 2015;82:540-51.
  20. Li L, Qin L, Lu HL, Li PJ, Song YJ, Yang RL. Methylene blue improves streptozotocin-induced memory deficit by restoring mitochondrial function in rats. Brain Res 2017;1657:208-14.
  21. Pakavathkumar P, Sharma G, Kaushal V, Foveau B, LeBlanc AC. Methylene blue inhibits caspases by oxidation of the catalytic cysteine. Sci Rep 2015;5:13730.
  22. Akoury E, Pickhardt M, Gajda M, Biernat J, Mandelkow E, Zweckstetter M. Mechanistic basis of phenothiazine-driven inhibition of Tau aggregation. Angew Chem Int Ed Engl 2013;52:3511-5.
  23. Crowe A, James MJ, Lee VM, Smith AB 3rd, Trojanowski JQ, Ballatore C, et al. Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation. J Biol Chem 2013;288:11024-37.
  24. Guo H, Albrecht S, Bourdeau M, Petzke T, Bergeron C, LeBlanc AC. Active caspase-6 and caspase-6- cleaved tau in neuropil threads, neuritic plaques, and neurofibrillary tangles of Alzheimer's disease. Am J Pathol 2004;165:523-31.
  25. Albrecht S, Bourdeau M, Bennett D, Mufson EJ, Bhattacharjee M, LeBlanc AC. Activation of caspase-6 in aging and mild cognitive impairment. Am J Pathol 2007;170:1200-9.
  26. Albrecht S, Bogdanovic N, Ghetti B, Winblad B, LeBlanc AC. Caspase-6 activation in familial alzheimer disease brains carrying amyloid precursor protein or presenilin I or presenilin II mutations. J Neuropathol Exp Neurol 2009;68:1282-93.
  27. Ramcharitar J, Afonso VM, Albrecht S, Bennett DA, LeBlanc AC. Caspase-6 activity predicts lower episodic memory ability in aged individuals. Neurobiol Aging 2013;34:1815-24.
  28. LeBlanc AC, Ramcharitar J, Afonso V, Hamel E, Bennett DA, Pakavathkumar P, et al. Caspase-6 activity in the CA1 region of the hippocampus induces age-dependent memory impairment. Cell Death Differ 2014;21:696-706.
  29. Schirmer RH, Adler H, Pickhardt M, Mandelkow E. Lest we forget you--methylene blue.... Neurobiol Aging 2011;32:2325.e7-16.
  30. Goedert M, Spillantini MG. A century of Alzheimer's disease. Science 2006;314:777-81.
  31. Wischik CM, Edwards PC, Lai RY, Roth M, Harrington CR. Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc Natl Acad Sci U S A 1996;93:11213-8.
  32. Yarza R, Vela S, Solas M, Ramirez MJ. c-Jun N-terminal Kinase (JNK) Signaling as a Therapeutic Target for Alzheimer's Disease. Front Pharmacol 2016;6:321.
  33. Piedrahita D, Hernández I, López-Tobón A, Fedorov D, Obara B, Manjunath BS, et al. Silencing of CDK5 reduces neurofibrillary tangles in transgenic alzheimer's mice. J Neurosci 2010;30:13966-76.
  34. Hooper C, Killick R, Lovestone S. The GSK3 hypothesis of Alzheimer's disease. J Neurochem 2008;104:1433-9.
  35. Mietelska-Porowska A, Wasik U, Goras M, Filipek A, Niewiadomska G. Tau protein modifications and interactions: their role in function and dysfunction. Int J Mol Sci 2014;15:4671-713.
  36. Rodríguez-Martín T, Cuchillo-Ibáñez I, Noble W, Nyenya F, Anderton BH, Hanger DP. Tau phosphorylation affects its axonal transport and degradation. Neurobiol Aging 2013;34:2146-57.
  37. Sulistio YA, Heese K. The Ubiquitin-proteasome system and molecular chaperone deregulation in Alzheimer's disease. Mol Neurobiol 2016;53:905-31.
  38. Hattori M, Sugino E, Minoura K, In Y, Sumida M, Taniguchi T, et al. Different inhibitory response of cyanidin and methylene blue for filament formation of tau microtubule-binding domain. Biochem Biophys Res Commun 2008;374:158-63.
  39. Taniguchi S, Suzuki N, Masuda M, Hisanaga S, Iwatsubo T, Goedert M, et al. Inhibition of heparin-induced tau filament formation by phenothiazines, polyphenols, and porphyrins. J Biol Chem 2005;280:7614-23.
  40. O'Leary JC 3rd, Li Q, Marinec P, Blair LJ, Congdon EE, Johnson AG, et al. Phenothiazine-mediated rescue of cognition in tau transgenic mice requires neuroprotection and reduced soluble tau burden. Mol Neurodegener 2010;5:45.
  41. van Bebber F, Paquet D, Hruscha A, Schmid B, Haass C. Methylene blue fails to inhibit Tau and polyglutamine protein dependent toxicity in zebrafish. Neurobiol Dis 2010;39:265-71.
  42. Congdon EE, Wu JW, Myeku N, Figueroa YH, Herman M, Marinec PS, et al. Methylthioninium chloride (methylene blue) induces autophagy and attenuates tauopathy in vitro and in vivo. Autophagy 2012;8:609-22.
  43. Melis V, Magbagbeolu M, Rickard JE, Horsley D, Davidson K, Harrington KA, et al. Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models. Behav Pharmacol 2015;26:353-68.
  44. Hosokawa M, Arai T, Masuda-Suzukake M, Nonaka T, Yamashita M, Akiyama H, et al. Methylene blue reduced abnormal tau accumulation in P301L tau transgenic mice. PLoS One 2012;7:e52389.
  45. Baddeley TC, McCaffrey J, Storey JM, Cheung JK, Melis V, Horsley D, et al. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer's disease. J Pharmacol Exp Ther 2015;352:110-8.
  46. Wischik CM, Staff RT, Wischik DJ, Bentham P, Murray AD, Storey JM, et al. Tau aggregation inhibitor therapy: an exploratory phase 2 study in mild or moderate Alzheimer's disease. J Alzheimers Dis 2015;44:705-20.
  47. Gauthier S, Feldman HH, Schneider LS, Wilcock GK, Frisoni GB, Hardlund JH, et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer's disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet 2016;388:2873-84.
  48. NHS, Description of IBD, Available at: https://www. nhs.uk/conditions/inflammatory-bowel-disease/
  49. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol 2015;12:720-7.
  50. Kim DH, Cheon JH. Pathogenesis of Inflammatory Bowel Disease and Recent Advances in Biologic Therapies. Immune Netw 2017;17:25-40.
  51. Pirogov SS, Sokolov VV, Kaprin AD, Volchenko NN, Karpova ES, Pavlov PV, et al. Endocytoscopy--novel endoscopic diagnostics approach: Principles and procedure. Eksp Klin Gastroenterol 2015;4:12-21.
  52. Ichimasa K, Kudo SE, Mori Y, Wakamura K, Ikehara N, Kutsukawa M, et al. Double staining with crystal violet and methylene blue is appropriate for colonic endocytoscopy: an in vivo prospective pilot study. Dig Endosc 2014;26:403-8.
  53. Meining A. Confocal laser endomicroscopy and endocytoscopy. UpToDate. Available at: https:// www.uptodate.com/contents/confocal-laserendomicroscopy-and-endocytoscopy
  54. Freire P, Figueiredo P, Cardoso R, Donato MM, Ferreira M, Mendes S, et al. Surveillance in ulcerative colitis: is chromoendoscopy-guided endomicroscopy always better than conventional colonoscopy? A randomized trial. Inflamm Bowel Dis 2014;20:2038-45.
  55. Mitsuyama K, Toyonaga A, Sasaki E, Ishida O, Ikeda H, Tsuruta O, et al. Soluble interleukin-6 receptors in inflammatory bowel disease: relation to circulating interleukin-6. Gut 1995;36:45-9.
  56. Reinisch W, Gasché C, Tillinger W, Wyatt J, Lichtenberger C, Willheim M, et al. Clinical relevance of serum interleukin-6 in Crohn's disease: single point measurements, therapy monitoring, and prediction of clinical relapse. Am J Gastroenterol 1999;94:2156-64.
  57. Li Y, de Haar C, Chen M, Deuring J, Gerrits MM, Smits R, et al. Disease-related expression of the IL6/STAT3/SOCS3 signalling pathway in ulcerative colitis and ulcerative colitis-related carcinogenesis. Gut 2010;59:227-35.
  58. Dinc S, Caydere M, Akgul G, Yenidogan E, Hücümenoglu S, Rajesh M. Methylene Blue Inhibits the Inflammatory Process of the Acetic Acid-Induced Colitis in Rat Colonic Mucosa. Int Surg 2015;100:1364-74.
  59. Kannan K, Jain SK. Oxidative stress and apoptosis. Pathophysiology 2000;7:153-63.
  60. El Sayed NS, Sayed AS. Protective effect of methylene blue on TNBS-induced colitis in rats mediated through the modulation of inflammatory and apoptotic signalling pathways. Arch Toxicol 2019;93:2927-42.