Chitosan: Comparison
Please note this is a comparison between Version 3 by Lily Guo and Version 2 by Camila Xu.

Chitosan is a random copolymer comprising d-glucosamine (the deacetylated ones) and N-acetyl-d-glucosamine units.

  • chitosan
  • chitosan oligosaccharides
  • gut microbiot
Please wait, diff process is still running!

References

  1. Thadathil, N.; Velappan, S.P. Recent developments in chitosanase research and its biotechnological applications: A review. Food Chem. 2014, 150, 392–399. [Google Scholar] [CrossRef] [PubMed]
  2. Guo, X.; Sun, T.; Zhong, R.; Ma, L.; You, C.; Tian, M.; Li, H.; Wang, C. Effects of Chitosan Oligosaccharides on Human Blood Components. Front. Pharmacol. 2018, 9, 1412. [Google Scholar] [CrossRef] [PubMed]
  3. Hao, C.; Wang, W.; Wang, S.; Zhang, L.; Guo, Y. An Overview of the Protective Effects of Chitosan and Acetylated Chitosan Oligosaccharides against Neuronal Disorders. Mar. Drugs 2017, 15, 89. [Google Scholar] [CrossRef] [PubMed]
  4. Jafari, H.; Bernaerts, K.V.; Dodi, G.; Shavandi, A. Chitooligosaccharides for wound healing biomaterials engineering. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 117, 111266. [Google Scholar] [CrossRef]
  5. Wang, W.; Xue, C.; Mao, X. Chitosan: Structural modification, biological activity and application. Int. J. Biol. Macromol. 2020, 164, 4532–4546. [Google Scholar] [CrossRef]
  6. Brás, T.; Rosa, D.; Gonçalves, A.C.; Gomes, A.C.; Alves, V.D.; Crespo, J.G.; Duarte, M.F.; Neves, L.A. Development of bioactive films based on chitosan and Cynara cardunculus leaves extracts for wound dressings. Int. J. Biol. Macromol. 2020, 163, 1707–1718. [Google Scholar] [CrossRef]
  7. Wu, X.; Kim, M.J.; Yang, H.J.; Park, S. Chitosan alleviated menopausal symptoms and modulated the gut microbiota in estrogen-deficient rats. Eur. J. Nutr. 2020. [Google Scholar] [CrossRef]
  8. Sarkar, S.; Das, D.; Dutta, P.; Kalita, J.; Wann, S.B.; Manna, P. Chitosan: A promising therapeutic agent and effective drug delivery system in managing diabetes mellitus. Carbohydr. Polym. 2020, 247, 116594. [Google Scholar] [CrossRef]
  9. Belorkar, S.A.; Gupta, A.K. Oligosaccharides: A boon from nature’s desk. AMB Express 2016, 6, 82. [Google Scholar] [CrossRef]
  10. Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef] [PubMed]
  11. Yang, C.M.; Ferket, P.R.; Hong, Q.H.; Zhou, J.; Cao, G.T.; Zhou, L.; Chen, A.G. Effect of chito-oligosaccharide on growth performance, intestinal barrier function, intestinal morphology and cecal microflora in weaned pigs. J. Anim. Sci. 2012, 90, 2671–2676. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, C.; Jiao, S.; Wang, Z.A.; Du, Y. Exploring Effects of Chitosan Oligosaccharides on Mice Gut Microbiota in in vitro Fermentation and Animal Model. Front. Microbiol. 2018, 9, 2388. [Google Scholar] [CrossRef] [PubMed]
  13. Dehghan, P.; Pourghassem Gargari, B.; Asghari Jafar-abadi, M. Oligofructose-enriched inulin improves some inflammatory markers and metabolic endotoxemia in women with type 2 diabetes mellitus: A randomized controlled clinical trial. Nutrition 2014, 30, 418–423. [Google Scholar] [CrossRef] [PubMed]
  14. Guan, G.; Azad, M.A.K.; Lin, Y.; Kim, S.W.; Tian, Y.; Liu, G.; Wang, H. Biological Effects and Applications of Chitosan and Chito-Oligosaccharides. Front. Physiol. 2019, 10, 516. [Google Scholar] [CrossRef] [PubMed]
  15. Patterson, E.; Ryan, P.M.; Cryan, J.F.; Dinan, T.G.; Ross, R.P.; Fitzgerald, G.F.; Stanton, C. Gut microbiota, obesity and diabetes. Postgrad. Med. J. 2016, 92, 286–300. [Google Scholar] [CrossRef] [PubMed]
  16. Weiss, G.A.; Hennet, T. Mechanisms and consequences of intestinal dysbiosis. Cell. Mol. Life Sci. CMLS 2017, 74, 2959–2977. [Google Scholar] [CrossRef]
  17. Derrien, M.; van Passel, M.W.; van de Bovenkamp, J.H.; Schipper, R.G.; de Vos, W.M.; Dekker, J. Mucin-bacterial interactions in the human oral cavity and digestive tract. Gut Microbes 2010, 1, 254–268. [Google Scholar] [CrossRef]
  18. Johansson, M.E.; Phillipson, M.; Petersson, J.; Velcich, A.; Holm, L.; Hansson, G.C. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl. Acad. Sci. USA 2008, 105, 15064–15069. [Google Scholar] [CrossRef]
  19. Ottman, N.; Davids, M.; Suarez-Diez, M.; Boeren, S.; Schaap, P.J.; Martins Dos Santos, V.A.P.; Smidt, H.; Belzer, C.; de Vos, W.M. Genome-Scale Model and Omics Analysis of Metabolic Capacities of Akkermansia muciniphila Reveal a Preferential Mucin-Degrading Lifestyle. Appl. Environ. Microbiol. 2017, 83, e01014-17. [Google Scholar] [CrossRef]
  20. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [PubMed]
  21. Macchione, I.G.; Lopetuso, L.R.; Ianiro, G.; Napoli, M.; Gibiino, G.; Rizzatti, G.; Petito, V.; Gasbarrini, A.; Scaldaferri, F. Akkermansia muciniphila: Key player in metabolic and gastrointestinal disorders. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8075–8083. [Google Scholar] [PubMed]
  22. Schneeberger, M.; Everard, A.; Gómez-Valadés, A.G.; Matamoros, S.; Ramírez, S.; Delzenne, N.M.; Gomis, R.; Claret, M.; Cani, P.D. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci. Rep. 2015, 5, 16643. [Google Scholar] [CrossRef] [PubMed]
  23. Miura, K.; Ohnishi, H. Role of gut microbiota and Toll-like receptors in nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 7381–7391. [Google Scholar] [CrossRef]
  24. Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation 2016, 133, 2434–2446. [Google Scholar] [CrossRef]
  25. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [Google Scholar] [CrossRef]
  26. Levin, P.D.; Weissman, C. Obesity, metabolic syndrome, and the surgical patient. Med. Clin. N. Am. 2009, 93, 1049–1063. [Google Scholar] [CrossRef]
  27. McCracken, E.; Monaghan, M.; Sreenivasan, S. Pathophysiology of the metabolic syndrome. Clin. Dermatol. 2018, 36, 14–20. [Google Scholar] [CrossRef]
  28. Jameson, J.L. Endocrinology: Adult and Pediatric, 7th ed.; Elsevier Saunders: Philadelphia, PA, USA, 2016. [Google Scholar]
  29. Canfora, E.E.; Meex, R.C.R.; Venema, K.; Blaak, E.E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nature reviews. Endocrinology 2019, 15, 261–273. [Google Scholar]
  30. Dong, T.S.; Jacobs, J.P. Nonalcoholic fatty liver disease and the gut microbiome: Are bacteria responsible for fatty liver? Exp. Biol. Med. 2019, 244, 408–418. [Google Scholar] [CrossRef] [PubMed]
  31. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed]
  32. Samson, S.L.; Garber, A.J. Metabolic syndrome. Endocrinol. Metab. Clin. North Am. 2014, 43, 1–23. [Google Scholar] [CrossRef]
  33. De la Cuesta-Zuluaga, J.; Mueller, N.T.; Corrales-Agudelo, V.; Velásquez-Mejía, E.P.; Carmona, J.A.; Abad, J.M.; Escobar, J.S. Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short-Chain Fatty Acid-Producing Microbiota in the Gut. Diabetes Care 2017, 40, 54–62. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, G.S.; Hoyte, C. Review of Biguanide (Metformin) Toxicity. J. Intensive Care Med. 2019, 34, 863–876. [Google Scholar] [CrossRef]
  35. Kim, J.; Lee, H.; An, J.; Song, Y.; Lee, C.K.; Kim, K.; Kong, H. Alterations in Gut Microbiota by Statin Therapy and Possible Intermediate Effects on Hyperglycemia and Hyperlipidemia. Front. Microbiol. 2019, 10, 1947. [Google Scholar] [CrossRef]
  36. Khan, T.J.; Ahmed, Y.M.; Zamzami, M.A.; Siddiqui, A.M.; Khan, I.; Baothman, O.A.S.; Mehanna, M.G.; Kuerban, A.; Kaleemuddin, M.; Yasir, M. Atorvastatin Treatment Modulates the Gut Microbiota of the Hypercholesterolemic Patients. Omics J. Integr. Biol. 2018, 22, 154–163. [Google Scholar] [CrossRef]
  37. Li, S.; Yu, Y.; Jin, Z.; Dai, Y.; Lin, H.; Jiao, Z.; Ma, G.; Cai, W.; Han, B.; Xiang, X. Prediction of pharmacokinetic drug-drug interactions causing atorvastatin-induced rhabdomyolysis using physiologically based pharmacokinetic modelling. Biomed. Pharmacother. Biomed. Pharmacother. 2019, 119, 109416. [Google Scholar] [CrossRef]
  38. Watanabe, K.; Oda, S.; Matsubara, A.; Akai, S.; Yokoi, T. Establishment and characterization of a mouse model of rhabdomyolysis by coadministration of statin and fibrate. Toxicol. Lett. 2019, 307, 49–58. [Google Scholar] [CrossRef]
  39. Wang, Z.; Zhang, F.; Yan, Y.; Zhang, Z.; Wang, L.; Qin, C. Lipid-lowering activities of chitosan and its quaternary ammonium salt for the hyperlipidemia rats induced by high-fat diets. Int. J. Biol. Macromol. 2019, 132, 922–928. [Google Scholar] [CrossRef]
  40. Luan, F.; Wei, L.; Zhang, J.; Tan, W.; Chen, Y.; Dong, F.; Li, Q.; Guo, Z. Preparation and Characterization of Quaternized Chitosan Derivatives and Assessment of Their Antioxidant Activity. Molecules 2018, 23, 516. [Google Scholar] [CrossRef]
  41. Guan, G.; Wang, H.; Chen, S.; Liu, G.; Xiong, X.; Tan, B.; Duraipandiyan, V.; Al-Dhabi, N.A.; Fang, J. Dietary Chitosan Supplementation Increases Microbial Diversity and Attenuates the Severity of Citrobacter rodentium Infection in Mice. Mediat. Inflamm. 2016, 2016, 9236196. [Google Scholar] [CrossRef] [PubMed]
  42. Huang, L.; Chen, J.; Cao, P.; Pan, H.; Ding, C.; Xiao, T.; Zhang, P.; Guo, J.; Su, Z. Anti-obese effect of glucosamine and chitosan oligosaccharide in high-fat diet-induced obese rats. Mar. Drugs 2015, 13, 2732–2756. [Google Scholar] [CrossRef] [PubMed]
  43. Aguilar, A.; Zein, N.; Harmouch, E.; Hafdi, B.; Bornert, F.; Offner, D.; Clauss, F.; Fioretti, F.; Huck, O.; Benkirane-Jessel, N.; et al. Application of Chitosan in Bone and Dental Engineering. Molecules 2019, 24, 3009. [Google Scholar] [CrossRef] [PubMed]
  44. Bravo-Anaya, L.M.; Soltero, J.F.; Rinaudo, M. DNA/chitosan electrostatic complex. Int. J. Biol. Macromol. 2016, 88, 345–533. [Google Scholar] [CrossRef]
  45. Ahmed, K.F.; Aschi, A.; Nicolai, T. Formation and characterization of chitosan-protein particles with fractal whey protein aggregates. Colloids Surf. B Biointerfaces 2018, 196, 257–264. [Google Scholar] [CrossRef]
  46. Liu, S.H.; Chiu, C.Y.; Shi, C.M.; Chiang, M.T. Functional Comparison of High and Low Molecular Weight Chitosan on Lipid Metabolism and Signals in High-Fat Diet-Fed Rats. Mar. Drugs 2018, 16, 251. [Google Scholar] [CrossRef]
  47. Dimzon, I.K.; Ebert, J.; Knepper, T.P. The interaction of chitosan and olive oil: Effects of degree of deacetylation and degree of polymerization. Carbohydr. Polym. 2013, 92, 564–570. [Google Scholar] [CrossRef]
  48. Mohan, K.; Ganesan, A.R.; Muralisankar, T.; Jayakumar, R.; Sathishkumar, P.; Uthayakumar, V.; Chandirasekar, R.; Revathi, N. Recent insights into the extraction, characterization, and bioactivities of chitin and chitosan from insects. Trends Food Sci. Technol. 2020, 105, 17–42. [Google Scholar] [CrossRef]
  49. Michos, E.D.; McEvoy, J.W.; Blumenthal, R.S. Lipid Management for the Prevention of Atherosclerotic Cardiovascular Disease. N. Engl. J. Med. 2019, 16, 1557–1567. [Google Scholar] [CrossRef]
  50. [35]
  51. Rizzo, M.; Giglio, R.V.; Nikolic, D.; Patti, A.M.; Campanella, C.; Cocchi, M.; Katsiki, N.; Montalto, G. Effects of chitosan on plasma lipids and lipoproteins: A 4-month prospective pilot study. Angiology 2014, 65, 538–542. [Google Scholar] [CrossRef]
  52. Lopez-Santamarina, A.; Mondragon, A.D.C.; Lamas, A.; Miranda, J.M.; Franco, C.M.; Cepeda, A. Animal-Origin Prebiotics Based on Chitin: An Alternative for the Future? A Critical Review. Foods 2020, 9, 782. [Google Scholar] [CrossRef] [PubMed]
  53. Hardie, D.G. The AMP-activated protein kinase pathway--new players upstream and downstream. J. Cell Sci. 2004, 117, 5479–5487. [Google Scholar] [CrossRef] [PubMed]
  54. Clarke, P.R.; Hardie, D.G. Regulation of HMG-CoA reductase: Identification of the site phosphorylated by the AMP-activated protein kinase in vitro and in intact rat liver. EMBO J. 1990, 9, 2439–2446. [Google Scholar] [CrossRef] [PubMed]
  55. Srivastava, R.A.; Pinkosky, S.L.; Filippov, S.; Hanselman, J.C.; Cramer, C.T.; Newton, R.S. AMP-activated protein kinase: An emerging drug target to regulate imbalances in lipid and carbohydrate metabolism to treat cardio-metabolic diseases. J. Lipid Res. 2012, 53, 2490–2514. [Google Scholar] [CrossRef] [PubMed]
  56. Horton, J.D.; Goldstein, J.L.; Brown, M.S. SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Investig. 2002, 109, 1125–1131. [Google Scholar] [CrossRef]
  57. Ge, H.; Yang, G.; Huang, L.; Motola, D.L.; Pourbahrami, T.; Li, C. Oligomerization and regulated proteolytic processing of angiopoietin-like protein 4. J. Biol. Chem. 2004, 279, 2038–2045. [Google Scholar] [CrossRef]
  58. Lei, X.; Shi, F.; Basu, D.; Huq, A.; Routhier, S.; Day, R.; Jin, W. Proteolytic processing of angiopoietin-like protein 4 by proprotein convertases modulates its inhibitory effects on lipoprotein lipase activity. J. Biol. Chem. 2011, 286, 15747–15756. [Google Scholar] [CrossRef]
  59. Yin, W.; Romeo, S.; Chang, S.; Grishin, N.V.; Hobbs, H.H.; Cohen, J.C. Genetic variation in ANGPTL4 provides insights into protein processing and function. J. Biol. Chem. 2009, 284, 13213–13222. [Google Scholar] [CrossRef]
  60. Ge, H.; Cha, J.Y.; Gopal, H.; Harp, C.; Yu, X.; Repa, J.J.; Li, C. Differential regulation and properties of angiopoietin-like proteins 3 and 4. J. Lipid Res. 2005, 46, 1484–1490. [Google Scholar] [CrossRef]
  61. Davies, B.S.; Beigneux, A.P.; Barnes, R.H., 2nd; Tu, Y.; Gin, P.; Weinstein, M.M.; Nobumori, C.; Nyrén, R.; Goldberg, I.; Olivecrona, G.; et al. GPIHBP1 is responsible for the entry of lipoprotein lipase into capillaries. Cell Metab. 2010, 12, 42–52. [Google Scholar] [CrossRef]
  62. Kersten, S.; Lichtenstein, L.; Steenbergen, E.; Mudde, K.; Hendriks, H.F.; Hesselink, M.K.; Schrauwen, P.; Müller, M. Caloric restriction and exercise increase plasma ANGPTL4 levels in humans via elevated free fatty acids. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 969–974. [Google Scholar] [CrossRef]
  63. González-Muniesa, P.; de Oliveira, C.; Pérez de Heredia, F.; Thompson, M.P.; Trayhurn, P. Fatty acids and hypoxia stimulate the expression and secretion of the adipokine ANGPTL4 (angiopoietin-like protein 4/fasting-induced adipose factor) by human adipocytes. J. Nutr. Nutr. 2011, 4, 146–153. [Google Scholar] [CrossRef] [PubMed]
  64. Peng, H.; Chiu, T.Y.; Liang, Y.J.; Lee, C.J.; Liu, C.S.; Suen, C.S.; Yen, J.J.; Chen, H.T.; Hwang, M.J.; Hussain, M.M.; et al. PRAP1 is a novel lipid binding protein that promotes lipid absorption by facilitating MTTP-mediated lipid transport. J. Biol. Chem. 2020. [Google Scholar] [CrossRef] [PubMed]
  65. Xie, Y.; Matsumoto, H.; Kennedy, S.; Newberry, E.P.; Moritz, W.; DeBosch, B.J.; Moley, K.H.; Rubin, D.C.; Warner, B.W.; Kau, A.L.; et al. Impaired Chylomicron Assembly Modifies Hepatic Metabolism Through Bile Acid-Dependent and Transmissible Microbial Adaptations. Hepatology 2019, 70, 1168–1184. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, Q.; Vijayakumar, A.; Kahn, B.B. Metabolites as regulators of insulin sensitivity and metabolism. Nature reviews. Mol. Cell Biol. 2018, 19, 654–672. [Google Scholar]
  67. American Diabetes Association. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care 2019, 42 (Suppl. 1), S13–S28. [Google Scholar] [CrossRef]
  68. Jeong, S.; Min Cho, J.; Kwon, Y.I.; Kim, S.C.; Yeob Shin, D.; Ho Lee, J. Chitosan oligosaccharide (GO2KA1) improves postprandial glycemic response in subjects with impaired glucose tolerance and impaired fasting glucose and in healthy subjects: A crossover, randomized controlled trial. Nutr. Diabetes 2019, 9, 31. [Google Scholar] [CrossRef]
  69. Hers, I.; Vincent, E.E.; Tavaré, J.M. Akt signalling in health and disease. Cell. Signal. 2011, 23, 1515–1527. [Google Scholar] [CrossRef]
  70. Liu, T.Y.; Shi, C.X.; Gao, R.; Sun, H.J.; Xiong, X.Q.; Ding, L.; Chen, Q.; Li, Y.H.; Wang, J.J.; Kang, Y.M.; et al. Irisin inhibits hepatic gluconeogenesis and increases glycogen synthesis via the PI3K/Akt pathway in type 2 diabetic mice and hepatocytes. Clin. Sci. 2015, 129, 839–850. [Google Scholar] [CrossRef]
  71. Zhang, Y.; Chen, J.; Zeng, Y.; Huang, D.; Xu, Q. Involvement of AMPK activation in the inhibition of hepatic gluconeogenesis by Ficus carica leaf extract in diabetic mice and HepG2 cells. Biomed. Pharmacother. Biomed. Pharmacother. 2019, 109, 188–194. [Google Scholar] [CrossRef]
  72. Liu, S.H.; Chang, Y.H.; Chiang, M.T. Chitosan reduces gluconeogenesis and increases glucose uptake in skeletal muscle in streptozotocin-induced diabetic rats. J. Agric. Food Chem. 2010, 58, 5795–5800. [Google Scholar] [CrossRef]
  73. Catrysse, L.; van Loo, G. Inflammation and the Metabolic Syndrome: The Tissue-Specific Functions of NF-κB. Trends Cell Biol. 2017, 27, 417–429. [Google Scholar] [CrossRef] [PubMed]
  74. Kunanusornchai, W.; Witoonpanich, B.; Tawonsawatruk, T.; Pichyangkura, R.; Chatsudthipong, V.; Muanprasat, C. Chitosan oligosaccharide suppresses synovial inflammation via AMPK activation: An in vitro and in vivo study. Pharmacol. Res. 2016, 113, 458–467. [Google Scholar] [CrossRef] [PubMed]
  75. Rogers, M.A.M.; Aronoff, D.M. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome. Clin. Microbiol. Infect. 2016, 22, 178.e1–178.e9. [Google Scholar] [CrossRef] [PubMed]
  76. Muanprasat, C.; Chatsudthipong, V. Chitosan oligosaccharide: Biological activities and potential therapeutic applications. Pharmacol. Ther. 2017, 170, 80–97. [Google Scholar] [CrossRef] [PubMed]
  77. Hashemi Goradel, N.; Najafi, M.; Salehi, E.; Farhood, B.; Mortezaee, K. Cyclooxygenase-2 in cancer: A review. J. Cell. Physiol. 2019, 235, 5683–5699. [Google Scholar] [CrossRef] [PubMed]
  78. Tai, T.S.; Sheu, W.H.; Lee, W.J.; Yao, H.T.; Chiang, M.T. Effect of chitosan on plasma lipoprotein concentrations in type 2 diabetic subjects with hypercholesterolemia. Diabetes Care 2000, 23, 1703–1704. [Google Scholar] [CrossRef]
  79. Kim, H.J.; Ahn, H.Y.; Kwak, J.H.; Shin, D.Y.; Kwon, Y.I.; Oh, C.G.; Lee, J.H. The effects of chitosan oligosaccharide (GO2KA1) supplementation on glucose control in subjects with prediabetes. Food Funct. 2014, 5, 2662–2669. [Google Scholar] [CrossRef]
  80. Huang, H.; Zou, Y.; Chi, H. Quantitative assessment of the effects of chitosan intervention on blood pressure control. Drug Des. Dev. Ther. 2018, 12, 67–75. [Google Scholar] [CrossRef]
More
Video Production Service