Патогенетична роль кишкового дисбіозу в розвитку ожиріння та цукрового діабету 2 типу
DOI:
https://doi.org/10.24026/1818-1384.3(55).2016.77658Ключові слова:
кишкова мікробіота, дисбіоз, цукровий діабет 2 типу, ожирінняАнотація
У статті висвітлені сучасні погляди на роль кишкового дисбіозу в розвитку ожиріння та цукрового діабету 2 типу. Зокрема, ці метаболічні розлади чітко асоційовані із зниженням різноманіття кишечної мікробіоти та підвищенням числа штамів, що найбільш ефективно добувають енергію з продуктів харчування. Такі відмінності у складі кишкової мікробіоти можуть бути застосовані як ранні діагностичні маркери розвитку цукрового діабету 2 типу у пацієнтів підвищеного ризику. Трансплантація кишкової мікробіоти від худих чоловічих донорів реципієнтам з метаболічним синдромом приводить до значного поліпшення чутливості до інсуліну в поєднанні з підвищенням різноманіття кишечної мікробіоти, в тому числі особливих бутират-продукуючих бактеріальних штамів, що володіють позитивними метаболічними ефектами.
Посилання
Ajslev, T.A. Childhood overweight after establishment of the gut microbiota: the role of delivery mode, pre-pregnancy weight and early administration of antibiotics. / T.A. Ajslev, C.S. Andersen, M. Gamborg, T.I.A. Sørensen, T. Jess // Int J Obes (Lond). – 2011. – 35. – P. 522–529. https://doi.org/10.1038/ijo.2011.27
Arumugam, M. MetaHIT Consortium. Enterotypes of the human gut microbiome / M. Arumugam, J. Raes, E. Pelletier, et al. // Nature. – 2011. – 473. – P. 174–180. https://doi.org/10.1038/nature09944
Backhed, F. The gut microbiota as an environmental factor that regulates fat storage / F. Backhed, H. Ding, T. Wang, et al. // Proc Natl Acad Sci USA. – 2004. – 101. – P. 15718–15723. https://doi.org/10.1073/pnas.0407076101
Blaser, M. Antibiotic overuse: stop the killing of beneficial bacteria / M. Blaser // Nature. – 2011. – 476. – P. 393–394. https://doi.org/10.1038/476393a
Cani, P.D. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia / P.D. Cani, A.M. Neyrinck, F. Fava, et al. // Diabetologia. – 2007. – 50. – P. 2374–2383. https://doi.org/10.1007/s00125-007-0791-0
Cani, P.D. Metabolic endotoxemia initiates obesity and insulin resistance / P.D. Cani, J. Amar, M.A. Iglesias, et al. // Diabetes. – 2007. – 56. – P. 1761–1772. https://doi.org/10.2337/db06-1491
Cho, I. Antibiotics in early life alter the murine colonic microbiome and adiposity / I. Cho, S. Yamanishi, L. Cox, et al. // Nature. – 2012. – 488. – P. 621–626. https://doi.org/10.1038/nature11400
Claesson, M.J. Evaluating the latest high-throughput molecular techniques for the exploration of microbial gut communities / M.J. Claesson, P.W. O’Toole // Gut Microbes. – 2010. – 1. – P. 277–278. https://doi.org/10.4161/gmic.1.4.12306
Cotillard, A. ANR MicroObes consortium. Dietary intervention impact on gut microbial gene richness / A. Cotillard, S.P. Kennedy, L.C. Kong, et al. // Nature. – 2013. – 500. – P. 585–588. https://doi.org/10.1038/nature12480
Cummings, J.H. Short chain fatty acids in the human colon / J.H. Cummings // Gut. – 1981. – 22. – P. 763–779. https://doi.org/10.1136/gut.22.9.763
Davie, J.R. Inhibition of histone deacetylase activity by butyrate / J.R. Davie // J Nutr. – 2003. – 133(Suppl.). – 2485S–2493S.
Danaei, G. Global Burden of Metabolic Risk Factors of Chronic Diseases Collaborating Group (Blood Glucose). National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2,7 million participants / G. Danaei, M.M. Finucane, Y. Lu, et al. // Lancet. – 2011. – 378. – P. 31–40. https://doi.org/10.1016/s0140-6736(11)60679-x
Defrancesco, M. Psychosocial state after bariatric surgery is associated with the serotonin-transporter promoter polymorphism / M. Defrancesco, J. Liebaert, G. Kemmler, et al. // Eat Weight Disord. – 2013. – 18. – P. 311–316. https://doi.org/10.1007/s40519-013-0045-8
Delaere, F. The role of sodium-coupled glucose co-transporter 3 in the satiety effect of portal glucose sensing / F. Delaere, A. Duchampt, L. Mounien, et al. // Mol Metab. – 2012. – 2. – P. 47–53. https://doi.org/10.1016/j.molmet.2012.11.003
Dethlefsen, L. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation / L. Dethlefsen, D.A. Relman // Proc Natl Acad Sci USA. – 2011. – 108 (Suppl. 1). – P. 4554–4561. https://doi.org/10.1073/pnas.1000087107
De Vadder, F. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits / F. De Vadder, P. Kovatcheva-Datchary, D. Goncalves, et al. // Cell. – 2014. – 156. – P. 84–96. https://doi.org/10.1016/j.cell.2013.12.016
Di Sabatino, A. Oral butyrate for mildly to moderately active Crohn’s disease / A. Di Sabatino, R. Morera, R. Ciccocioppo, et al. // Aliment Pharmacol Ther. – 2005. – 22. – P. 789–794. https://doi.org/10.1111/j.1365-2036.2005.02639.x
Donohoe, D.R. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon / D.R. Donohoe, N. Garge, X. Zhang, et al. // Cell Metab. – 2011. – 13. – P. 517–526. https://doi.org/10.1016/j.cmet.2011.02.018
Duncan, S.H. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces / S.H. Duncan, A. Belenguer, G. Holtrop, A.M. Johnstone, H.J. Flint, G.E. Lobley // Appl Environ Microbiol. – 2007. – 73. – P. 1073–1078. https://doi.org/10.1128/aem.02340-06
Fleissner, C.K. Absence of intestinal microbiota does not protect mice from diet-induced obesity / C.K. Fleissner, N. Huebel, M.M. Abd El-Bary, G. Loh, S. Klaus, M. Blaut // Br J Nutr. – 2010. – 104. – P. 919–929. https://doi.org/10.1017/s0007114510001303
Flint, H.J. Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis / H.J. Flint, E.A. Bayer, M.T. Rincon, R. Lamed, B.A. White // Nat Rev Microbiol. – 2008. – 6. – P. 121–131. https://doi.org/10.1038/nrmicro1817
Furet, J.-P. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers / J.-P. Furet, L.-C. Kong, J. Tap, et al. // Diabetes. – 2010. – 59. – P. 3049–3057. https://doi.org/10.2337/db10-0253
Gao, Z. Butyrate improves insulin sensitivity and increases energy expenditure in mice / Z. Gao, J. Yin, J. Zhang, et al. // Diabetes. – 2009. – 58. – P. 1509–1517. https://doi.org/10.2337/db08-1637
Gill, R.K. Regulation of intestinal serotonin transporter expression via epigenetic mechanisms: role of HDAC2 / R.K. Gill, A. Kumar, P. Malhotra, et al. // Am J Physiol Cell Physiol. – 2013. – 304. – C334–C341. https://doi.org/10.1152/ajpcell.00361.2012
Hartstra, A. Insights Into the Role of the Microbiome in Obesity and Type 2 Diabetes / A. Hartstra, К. Bouter, F. Backhed, М. Nieuwdorp // Diabetes Care. – 2015. – 38. – P. 159–165. https://doi.org/10.2337/dc14-0769
Haub, S. Serotonin reuptake transporter (SERT) plays a critical role in the onset of fructose-induced hepatic steatosis in mice / S. Haub, G. Kanuri, V. Volynets, T. Brune, S.C. Bischoff, I. Bergheim // Am J Physiol Gastrointest Liver Physiol. – 2010. – 298. – G335–G344. https://doi.org/10.1152/ajpgi.00088.2009
Hildebrandt, M.A. High-fat diet determines the composition of the murine gut microbiome independently of obesity / M.A. Hildebrandt, C. Hoffmann, S.A. Sherrill-Mix, et al. // Gastroenterology. – 2009. – 137. – 1716–1724.e1–2. https://doi.org/10.1053/j.gastro.2009.08.042
Karlsson, F.H. Gut metagenome in European women with normal, impaired and diabetic glucose control / F.H. Karlsson, V. Tremaroli, I. Nookaew, et al. // Nature. – 2013. – 498. – P. 99–103. https://doi.org/10.1038/nature12198
Koopman, K.E. Diet-induced changes in the lean brain: hypercaloric high-fat-high-sugar snacking decreases serotonin transporters in the human hypothalamic region / K.E. Koopman, J. Booij, E. Fliers, M.J. Serlie, S.E. la Fleur // Mol Metab. – 2013. – 2. – P. 417–422. https://doi.org/10.1016/j.molmet.2013.07.006
Kootte, R.S. The therapeutic potential of manipulating gut microbiota in obesity and type 2 diabetes mellitus / R.S. Kootte, A. Vrieze, F. Holleman, et al. // Diabetes Obes Metab. – 2012. – 14. – P. 112–120. https://doi.org/10.1111/j.1463-1326.2011.01483.x
Lee, Y.K. Has the microbiota played a critical role in the evolution of the adaptive immune system? / Y.K. Lee, S.K. Mazmanian // Science. – 2010. – 330. – P. 1768–1773. https://doi.org/10.1126/science.1195568
Ley, R.E. Obesity alters gut microbial ecology / R.E. Ley, F. Backhed, P. Turnbaugh, C.A. Lozupone, R.D. Knight, J.I. Gordon // Proc Natl Acad Sci USA. – 2005. – 102. – 11070–11075. https://doi.org/10.1073/pnas.0504978102
Ley, R.E. Microbial ecology: human gut microbes associated with obesity/ R.E. Ley, P.J. Turnbaugh, S. Klein, J.I. Gordon // Nature. – 2006. – 444. – 1022–1023. https://doi.org/10.1038/4441022a
Liou, A.P. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity / A.P. Liou, M. Paziuk, J.-M. Luevano, S. Machineni, P.J. Turnbaugh, L.M. Kaplan // Sci Transl Med. – 2013. – 5. – 178ra41. https://doi.org/10.1126/scitranslmed.3005687
Meyer, D. The bifidogenic effect of inulin and oligofructose and its consequences for gut health / D. Meyer, M. Stasse-Wolthuis // Eur J Clin Nutr. – 2009. – 63. – 1277–1289. https://doi.org/10.1038/ejcn.2009.64
Beneficial effect of oral administration of Lactobacillus casei strain Shirota on insulin resistance in dietinduced obesity mice / Naito E., Yoshida Y., Makino K., et al. // J Appl Microbiol. – 2011. – 110. – 650–657. https://doi.org/10.1111/j.1365-2672.2010.04922.x
Palermo, A. Prevention of type 2 diabetes mellitus: is it feasible? / A. Palermo, D. Maggi, A.R. Maurizi, P. Pozzilli, R. Buzzetti // Diabetes Metab Res Rev. – 2014. – 30(Suppl. 1). – 4–12. https://doi.org/10.1002/dmrr.2513
Qin, J. A metagenome-wide association study of gut microbiota in type 2 diabetes / J. Qin, Y. Li, Z. Cai, et al. // Nature. – 2012. – 490. – 55–60. https://doi.org/10.1038/nature11450
Rastall, R.A. Modulation of the microbial ecology of the human colon by probiotics, prebiotics and synbiotics to enhance human health: an overview of enabling science and potential applications / R.A. Rastall, G.R. Gibson, H.S. Gill, et al. // FEMS Microbiol Ecol. – 2005. – 52. – 145–152. https://doi.org/10.1016/j.femsec.2005.01.003
Ratner, C. Cerebral markers of the serotonergic system in rat models of obesity and after Roux-en-Y gastric bypass / C. Ratner, A. Ettrup, M. Bueter, et al. // Obesity (Silver Spring). – 2012. – 20. – 2133–2141. https://doi.org/10.1038/oby.2012.75
Romanova, I.V. Neurobiologic changes in the hypothalamus associated with weight loss after gastric bypass / I.V. Romanova, E.J.B. Ramos, Y. Xu, et al. // J Am Coll Surg. – 2004. – 199. – 887–895. https://doi.org/10.1016/j.jamcollsurg.2004.07.013
Samuel, B.S. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41 / B.S. Samuel, A. Shaito, T. Motoike, et al. // Proc Natl Acad Sci USA. – 2008. – 105. – 16767–16772. https://doi.org/10.1073/pnas.0808567105
Scheppach, W. Effects of short chain fatty acids on gut morphology and function / W. Scheppach // Gut. – 1994. – 35(Suppl.). – S35–S38. https://doi.org/10.1136/gut.35.1_suppl.s35
Scheppach, W. Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis / W. Scheppach, H. Sommer, T. Kirchner, et al. // Gastroenterology. – 1992. – 103. – 51–56. https://doi.org/10.1016/0016-5085(92)91094-k
Schwiertz, A. Microbiota and SCFA in lean and overweight healthy subjects / A. Schwiertz, D. Taras, K. Schafer, et al. // Obesity (Silver Spring). – 2010. – 18. – 190–195. https://doi.org/10.1038/oby.2009.167
Simansky, K.J. Serotonergic control of the organization of feeding and satiety / K.J. Simansky // Behav Brain Res. – 1996. – 73. – 37–42. https://doi.org/10.1016/0166-4328(96)00066-6
Simren, M. Rome Foundation Committee. Intestinal microbiota infunctional bowel disorders: a Rome foundation report / M. Simren, G. Barbara, H.J. Flint, et al. // Gut. – 2013. – 62. – 159–176. https://doi.org/10.1136/gutjnl-2012-302167
Sjostrom, L. Swedish Obese Subjects Study Scientific Group. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery / L. Sjostrom, A.-K. Lindroos, M. Peltonen, et al. // N Engl J Med. –2004. – 351. – 2683–2693. https://doi.org/10.1056/nejmoa035622
Smits, L.P. Therapeutic potential of fecal microbiota transplantation / L.P. Smits, K.E.C. Bouter, W.M. de Vos, T.J. Borody, M. Nieuwdorp // Gastroenterology. – 2013. – 145. – 946–953. https://doi.org/10.1053/j.gastro.2013.08.058
Tazoe, H. Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions / H. Tazoe, Y. Otomo, I. Kaji, R. Tanaka, S.-I. Karaki, A. Kuwahara // J Physiol Pharmacol. – 2008. – 59(Suppl. 2). – 251–262.
Tilg, H. Gut microbiome, obesity, and metabolic dysfunction / H. Tilg, A. Kaser // J Clin Invest. – 2011. – 121. – 2126–2132. https://doi.org/10.1172/jci58109
Thuny, F. Vancomycin treatment of infective endocarditis is linked with recently acquired obesity / F. Thuny, H. Richet, J.-P. Casalta, E. Angelakis, G. Habib, D. Raoult // PLoS ONE. – 2010. – 5. – e9074. https://doi.org/10.1371/journal.pone.0009074
Tremaroli, V. Functional interactions between the gut microbiota and host metabolism / V. Tremaroli, F. B ̈ackhed // Nature. – 2012. – 489. – 242–249. https://doi.org/10.1038/nature11552
Turnbaugh, P.J. An obesity-associated gut microbiome with increased capacity for energy harvest / P.J. Turnbaugh, R.E. Ley, M.A. Mahowald, V. Magrini, E.R. Mardis, J.I. Gordon // Nature. – 2006. – 444. – 1027–1031. https://doi.org/10.1038/nature05414
Turnbaugh, P.J. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice / P.J. Turnbaugh, V.K. Ridaura, J.J. Faith, F.E.Rey, R. Knight, J.I. Gordon // Sci Transl Med. – 2009. – 1. – 6–14. https://doi.org/10.1126/scitranslmed.3000322
Vrieze, A. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome / A. Vrieze, E. Van Nood, F. Holleman, et al. // Gastroenterology. – 2012. – 143. – 913–916.e7. https://doi.org/10.1053/j.gastro.2012.06.031
Won, Y.-J. b-Hydroxybutyrate modulates N-type calcium channels in rat sympathetic neurons by acting as an agonist for the G-protein-coupled receptor FFA3 / Y.-J. Won, V.B. Lu, H.L. Puhl 3rd, S.R. Ikeda // J Neurosci. – 2013. – 33. – 19314–19325. https://doi.org/10.1523/jneurosci.3102-13.2013
Wu, G.D. Linking long-term dietary patterns with gut microbial enterotypes / G.D. Wu, J. Chen, C. Hoffmann, et al. // Science. – 2011. – 334. – 105–108. https://doi.org/10.1126/science.1208344
Yadav, H. Antidiabetic effect of probiotic dahi containing Lactobacillus acidophilus and Lactobacillus casei in high fructose fed rats / H. Yadav, S. Jain, P.R. Sinha // Nutrition. – 2007. – 23. – 62–68. https://doi.org/10.1016/j.nut.2006.09.002
Yamada, T. Serotonin stimulates endotoxin translocation via 5-HT3 receptors in the rat ileum / T. Yamada, A. Inui, N. Hayashi, M. Fujimura, M. Fujimiya // Am J Physiol Gastrointest Liver Physiol. – 2003. – 284. – G782–G788. https://doi.org/10.1152/ajpgi.00376.2002
Zhang, H. Human gut microbiota in obesity and after gastric bypass / H. Zhang, J.K. DiBaise, A. Zuccolo, et al. // Proc Natl Acad Sci USA. – 2009. – 106. – 2365–2370. https://doi.org/10.1073/pnas.0812600106
Zhu, B. Human gut microbiome: the second genome of human body / B. Zhu, X. Wang, L. Li // Protein Cell. – 2010. – 1. – 718–725. https://doi.org/10.1007/s13238-010-0093-z
##submission.downloads##
Опубліковано
Як цитувати
Номер
Розділ
Ліцензія
Авторське право (c) 2016 Клінічна ендокринологія та ендокринна хірургія
Ця робота ліцензується відповідно до Creative Commons Attribution-NonCommercial 4.0 International License.