Effectiveness of Experimental Colitis Therapy with Original Vitamin D3 Rectal Suppositories

M.V. Osikov, M.S. Boyko, A.A. Fedosov, M.A. Ilyinykh

 
International Journal of Biomedicine. 2022;12(1):124-133.
DOI: 10.21103/Article12(1)_OA13
Originally published March 10, 2022

Abstract: 

Background: Pathogenesis of inflammatory bowel disease (IBD) is insufficiently explored, while most of the therapeutic agents used for IBD cases have undesirable side effects, which restrict their administration.The aim of this research was to study the influence of vitamin D3 formulated into original rectal suppositories on the parameters of clinical score, morphology, and oxidative lipid and protein destruction in the colonic lesion in the cases of experimental colitis (EC). 
Methods and Results: The experiment was performed on 98 Wistar male rats weighing 210-230 g. EC condition was induced by two-phase administration (dermal application and per rectum) of 3% alcohol solution of oxazolone. Originator polyethylene glycol-based suppositories, which contained 1500 МЕ of vitamin D3, were administered per rectum every 12 hours. Clinical score was defined according to the Disease Activity Index (DAI) scale. Morphometry was run using the software program “ImageScope M” (Russia). Damage of the colonic tissue was estimated on Tissue Damage Index (TDI).
The following parameters were determined in the colonic lesion: neutrophil count (NC), lymphocyte count (LC), eosinophil count (EC), histiocyte count (HC), plasma cell count (PC), fibroblast count (FC), the diameter of the ulcerous defect, TDI, MPO expression, and TNF-α expression. The following parameters were determined in the damaged tissue homogenate: lipid peroxidation product count (LPP) and protein oxidative modification (POM) count.
In cases of oxazolone-induced EC, on Days 2, 4, and 6, we registered clinical and laboratory signs, an ulcerous defect in the damaged area of the colon, all of which are typical for IBD conditions. There was an increase in DAI (peak on Day 6) and TDI (peak on Day 2). We also found an increase in NC (peak on Day 2), LC (peak on Day 6), EC (peak on Day 2), PC (peak on Day 2), HC (peak on Day 2), and FC (peak on Day 2). There was an increase in MPO expression (peak on Day 2) and TNF-α expression (peak on Days 2 and 4). We observed increases in the primary, secondary, and end LPP counts and the early-phase and late-phase POM counts in spontaneous and induced modes.
An administration of 1500ME vitamin D3 rectal suppositories every 12 hours for 6 days decreased the severity of clinical manifestations and DAI. It reduced the area of the ulcerous defect and decreased the TDI on Days 4 and 6 of the experiment. On the background of using vitamin D3 rectal suppositories, we found a decrease in NC, EC, LC, and PC in the damaged area and an increase in HC and FC on Days 2, 4, and 6 from the start of the experiment. Administration of D3 rectal suppositories decreased МРО expression and TNF-α expression on Days 4 and 6 of EC. In the damaged area of the colon, we observed a decrease in the counts of the primary, secondary, and end LPP on Days 4 and 6 of the experiment. We also documented a decrease in the POM count in spontaneous mode on Day 2 and on Day 6 in induced mode.
Conclusion: Vitamin D3 as a constituent of originator rectal suppositories in total dose 18,000 МЕ in the pre-clinical phase of EC decreases the intensity of EC clinical manifestations. It reduces the count of the cells that take part in tissue destruction in the colonic wall, of TNF-α and MPO expression levels, and LPP- and POM product count. It increases the count of the cells, which promotes tissue reparation. The obtained results are essential for carrying out further research aimed at elaboration of the mechanism of the D3 effect in cases of IBD and at its possible clinical use.

Keywords: 
experimental colitis • rectal suppositories • vitamin D3 • oxidative stress
References: 
  1. Knyazev OV, Shkurko TV, Kagramanova AV, Veselov AV, Nikonov EL. [Epidemiology of inflammatory bowel disease. The current state of the problem (Literature review)]. Evidence-Based Gastroenterology. 2020;9(2):66–73. doi: 10.17116/dokgastro2020902166.  [Article in Russian].
  2. Mak WY, Zhao M, Ng SC, Burisch J. The epidemiology of inflammatory bowel disease: East meets west. J Gastroenterol Hepatol. 2020 Mar;35(3):380-389. doi: 10.1111/jgh.14872.
  3. Dolgushina AI, Khusainova GM, Vasilenko AG, Kononets VA. [The prevalence of inflammatory bowel disease in the Chelyabinsk region]. Almanac of Clinical Medicine. 2019;47(6):511-517.  doi:10.18786/2072-0505-2019-47-066. [Article in Russian].
  4. Tohari AM, Alhasani RH, Biswas L, Patnaik SR, Reilly J, Zeng Z, Shu X. Vitamin D Attenuates Oxidative Damage and Inflammation in Retinal Pigment Epithelial Cells. Antioxidants (Basel). 2019 Aug 24;8(9):341. doi: 10.3390/antiox8090341. 
  5. Kopecki Z, Yang G, Treloar S, Mashtoub S, Howarth GS, Cummins AG, Cowin AJ. Flightless I exacerbation of inflammatory responses contributes to increased colonic damage in a mouse model of dextran sulphate sodium-induced ulcerative colitis. Sci Rep. 2019 Sep 5;9(1):12792. doi: 10.1038/s41598-019-49129-6.
  6. Sitkin SI, Vakhitov TY, Demyanova EV. [The microbiome, colon dysbiosis, and inflammatory bowel disease: when function is more important than taxonomy]. Almanac of Clinical Medicine. 2018;46(5):396–425. doi:10.18786/2072-0505-2018-46-5-396-425. [Article in Russian].
  7. Ungaro R, Mehandru S, Allen PB, Peyrin-Biroulet L, Colombel JF. Ulcerative colitis. Lancet. 2017 Apr 29;389(10080):1756-1770. doi: 10.1016/S0140-6736(16)32126-2. 
  8. Yokoyama Y, Kamikozuru K, Nakamura S. Granulomonocytapheresis as a cell-based therapy in an ulcerative colitis patient complicated by aminosalicylate-induced severe lymphocytopenia and pneumonia. Cytotherapy. 2016 Sep;18(9):1234-6. doi: 10.1016/j.jcyt.2016.05.016. 
  9. Ivashkin VT, Shelygin YA, Khalif IL, Belousova EA. [Clinical recommendations of the Russian gastroenterological association and the association of coloproctologists of Russia for the diagnosis and treatment of ulcerative colitis]. Coloproctology. 2017;1(59):6-30. [Article in Russian].
  10. Šimoliūnas E, Rinkūnaitė I, Bukelskienė Ž, Bukelskienė V. Bioavailability of Different Vitamin D Oral Supplements in Laboratory Animal Model. Medicina (Kaunas). 2019 Jun 10;55(6):265. doi: 10.3390/medicina55060265.
  11. Yamamoto E, Jørgensen TN. Immunological effects of vitamin D and their relations to autoimmunity. J Autoimmun. 2019 Jun;100:7-16. doi: 10.1016/j.jaut.2019.03.002. 
  12. Bakke D, Sun J. Ancient Nuclear Receptor VDR With New Functions: Microbiome and Inflammation. Inflamm Bowel Dis. 2018 May 18;24(6):1149-1154. doi: 10.1093/ibd/izy092.
  13. Harrison SR, Li D, Jeffery LE, Raza K, Hewison M. Vitamin D, Autoimmune Disease and Rheumatoid Arthritis. Calcif Tissue Int. 2020 Jan;106(1):58-75. doi: 10.1007/s00223-019-00577-2.
  14. Liu J, Wang W, Liu K, Wan D, Wu Z, Cao Z, Luo Y, Xiao C, Yin M. Vitamin D receptor gene polymorphisms are associated with psoriasis susceptibility and the clinical response to calcipotriol in psoriatic patients. Exp Dermatol. 2020 Dec;29(12):1186-1190. doi: 10.1111/exd.14202. 
  15. Del Pinto R, Ferri C, Cominelli F. Vitamin D Axis in Inflammatory Bowel Diseases: Role, Current Uses and Future Perspectives. Int J Mol Sci. 2017 Nov 7;18(11):2360. doi: 10.3390/ijms18112360.
  16. Naderpoor N, Mousa A, Fernanda Gomez Arango L, Barrett HL, Dekker Nitert M, de Courten B. Effect of Vitamin D Supplementation on Faecal Microbiota: A Randomised Clinical Trial. Nutrients. 2019 Nov 27;11(12):2888. doi: 10.3390/nu11122888.
  17. Simonyan EV, Osikov MV, Boyko MS, Bakeeva AE. Remedy with vitamin D3 for the treatment of ulcerative colitis in the form of rectal suppositories. Patent RU, No. 2709209. 2019. Bull. #35.
  18. Osikov MV, Boyko MS, Simonyan EV, Ushakova VA. [Immunotropic effects of vitamin D3 in original rectal suppositories in experimental ulcerative colitis]. Medical Immunology (Russia). 2021;23(3):497-508. doi: 10.15789/1563-0625-IEO-2176. [Article in Russian].
  19. Directive 2010/63/EU of the European Parliament and of the Council of September 22, 2010, on the Protection of Animals Used for Scientific Purposes 2010. Official Journal of the European Union. L 276/33. Available from: https://eur-lex.europa.eu/eli/dir/2010/63/oj.
  20. Hoving JC, Keeton R, Höft MA, Ozturk M, Otieno-Odhiambo P, Brombacher F. IL-4 Receptor-Alpha Signalling of Intestinal Epithelial Cells, Smooth Muscle Cells, and Macrophages Plays a Redundant Role in Oxazolone Colitis. Mediators Inflamm. 2020 Jan 17;2020:4361043. doi: 10.1155/2020/4361043. 
  21. Osikov MV, Boyko MS, Simonyan EV. [The severity of the acute phase reaction to the use in experimental ulcerative colitis under D3 conditions in original rectal suppositories]. Pathological Physiology and Experiment. 2021;65(4):80–88. doi: 10.25557/0031-2991.2021.04.80-88. [Article in Russian].
  22. Kim JJ, Shajib MS, Manocha MM, Khan WI. Investigating intestinal inflammation in DSS-induced model of IBD. J Vis Exp. 2012 Feb 1;(60):3678. doi: 10.3791/3678.
  23. Yao J, Lu Y, Zhi M, Hu P, Wu W, Gao X. Dietary n‑3 polyunsaturated fatty acids ameliorate Crohn's disease in rats by modulating the expression of PPAR‑γ/NFAT. Mol Med Rep. 2017 Dec;16(6):8315-8322. doi: 10.3892/mmr.2017.7673. 
  24. Volchegorskiĭ IA, Nalimov AG, Iarovinskiĭ BG, Lifshits RI. [Comparison of various approaches to the determination of the products of lipid peroxidation in heptane-isopropanol extracts of blood]. Vopr Med Khim. 1989 Jan-Feb;35(1):127-31. [Article in Russian].
  25. Lvovskaya EI, Volchegorsky IA, Shemyakov SE, Lifshits RI. [Spectrophotometric determination of LPO end products]. Voprosy Meditsinskoi Khimii. 1991;4:92–93. [Article in Russian].
  26. Dubinina EE. Products of oxygen metabolism in the functional activity of cells (life and death, creation and destruction). Physiological and clinical-biochemical aspects. SPb.: "Medical press," 2006. [In Russian].
  27. Fomina MA. A method for a comprehensive assessment of the content of products of oxidative modification of proteins in tissues and biological fluids: Guidelines. Ryazan, 2014. [In Russian].
  28. Larabi A, Barnich N, Nguyen HTT. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy. 2020 Jan;16(1):38-51. doi: 10.1080/15548627.2019.1635384. 
  29. Lynch WD, Hsu R. Ulcerative Colitis. 2021 Jun 18. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan–. PMID: 29083748.
  30. Tian T, Wang Z, Zhang J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. Oxid Med Cell Longev. 2017;2017:4535194. doi: 10.1155/2017/4535194. 
  31. De Schepper S, Stakenborg N, Matteoli G, Verheijden S, Boeckxstaens GE. Muscularis macrophages: Key players in intestinal homeostasis and disease. Cell Immunol. 2018 Aug;330:142-150. doi: 10.1016/j.cellimm.2017.12.009. 
  32. Murdaca G, Tonacci A, Negrini S, Greco M, Borro M, Puppo F, Gangemi S. Emerging role of vitamin D in autoimmune diseases: An update on evidence and therapeutic implications. Autoimmun Rev. 2019 Sep;18(9):102350. doi: 10.1016/j.autrev.2019.102350. 
  33. Teixeira TM, da Costa DC, Resende AC, Soulage CO, Bezerra FF, Daleprane JB. Activation of Nrf2-Antioxidant Signaling by 1,25-Dihydroxycholecalciferol Prevents Leptin-Induced Oxidative Stress and Inflammation in Human Endothelial Cells. J Nutr. 2017 Apr;147(4):506-513. doi: 10.3945/jn.116.239475. 
  34. Fakhoury HMA, Kvietys PR, AlKattan W, Anouti FA, Elahi MA, Karras SN, Grant WB. Vitamin D and intestinal homeostasis: Barrier, microbiota, and immune modulation. J Steroid Biochem Mol Biol. 2020 Jun;200:105663. doi: 10.1016/j.jsbmb.2020.105663. 
  35. Charoenngam N, Holick MF. Immunologic Effects of Vitamin D on Human Health and Disease. Nutrients. 2020 Jul 15;12(7):2097. doi: 10.3390/nu12072097. 

Download Article
Received February 4, 2022.
Accepted March 7, 2022.
©2022 International Medical Research and Development Corporation.