The Značaj mikrobne fermentacije za dugoročne zdravstvene efekte ishrane bogate proteinima

  • Mirjana Rajilić-Stojanović Univerzitet u Beogradu – Tehnološko-metalurški fakultet, Katedra za biohemijsko inženjerstvo i biotehnologiju
Ključne reči: crevna mikrobiota, ishrana bogata proteinima, rak, kardiometabolički rizik, fermentacija

Sažetak


Proteini su značajni makronutrijenti sa nizom dokazanih pozitivnih efekata na zdravlje. Danas je ishrana bogata proteinima široko primenjivana, naročito u cilju dostizanja i održavanje zdrave telesne mase. Iako se povezuje sa pozitivnim zdravstvenim efektima, dugoročna ishrana bazirana na visokom sadržaju proteina je zapravo povezana sa više oboljenja i sa kraćim životnim vekom. Negativni zdravstveni efekti proteinskih dijeta su, makar delimično, posredovani aktivnošću mikrobnih metabolita dobijenih fermentacijom proteina. Naime, sve nesvarene komponente hrane se u debelom crevu fermentišu od strane crevne mikrobiote. S obzirom na to da su bočni ostaci aminokiselina hemijski raznovrsni, nesvareni proteini obezbeđuju složenu smešu supstrata za mikrobnu fermentaciju. Korišćenjem različitih aminokiselina, mikrobiota može da proizvede toksična, genotoksična i kancerogena jedinjenja, ali i metabolite koji ometaju signalizaciju insulina ili kompromituju kardiovaskularno zdravlje. Postoje naučni dokazi da mikrobni metaboliti više aminokiselina mogu doprineti razvoju kardiovaskularnih bolesti i raka, tj. bolesti koje su povezane sa ishranom bogatom proteinima. Iako su metabolički proizvodi mikrobiote u principu korisni i komplementarni ljudskom metabolizmu, kada je sastav ishrane neuravnotežen (npr. kada su proteini prisutni u prevelikoj količini) aktivnost mikrobiote se menja i dolazi do proizvodnje štetnih produkata. Imajući navedeno u vidu, neophodno je da se pri osmišljavanju zdrave ishrane u obzir uzme i aktivnost crevne mikrobiote.

Reference

Pasiakos SM, McLellan TM, Lieberman HR. The effects of protein supplements on muscle mass, strength, and aerobic and anaerobic power in healthy adults: a systematic review. Sport Med. 2015;45:111–31.

Cava E, Yeat NC, Mittendorfer B. Preserving healthy muscle during weight loss. Adv Nutr. 2017;8(3):511–9.

Halton TL, Hu FB. The effects of high protein diets on thermogenesis, satiety and weight loss: a critical review. J Am Coll Nutr. 2004;23(5):373–85.

Blom WAM, Lluch A, Stafleu A, Vinoy S, Holst JJ, Schaafsma G, et al. Effect of a high-protein breakfast on the postprandial ghrelin response. Am J Clin Nutr. 2006;83(2):211–20.

Kitts DD, Weiler K. Bioactive proteins and peptides from food sources. Applications of bioprocesses used in isolation and recovery. Curr Pharm Des. 2003;9(16):1309–23.

Colgrave ML, Dominik S, Tobin AB, Stockmann R, Simon C, Howitt CA, et al. Perspectives on future protein production. J Agric Food Chem. 2021;69(50):15076–83.

Wolever TMS, Mehling C. Long-term effect of varying the source or amount of dietary carbohydrate on postprandial plasma glucose, insulin, triacylglycerol, and free fatty acid concentrations in subjects with impaired glucose tolerance. Am J Clin Nutr. 2003;77(3):612–21.

Seidelmann SB, Claggett B, Cheng S, Henglin M, Shah A, Steffen LM, et al. Dietary carbohydrate intake and mortality: a prospective cohort study and meta-analysis. Lancet Public Health. 2018;3(9):e419–e428.

Delimaris I. Adverse effects associated with protein intake above the recommended dietary allowance for adults. Int Sch Res Not. 2013;2013.

Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164(3):337–40.

Rajilić-Stojanović M. Function of the microbiota. Best Pract Res Clin Gastroenterol. 2013;27(1):5–16.

Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev. 2014 Sep;38(5):996–1047.

Oren A, Garrity GM. Valid publication of the names of forty-two phyla of prokaryotes. Int J Syst Evol Microbiol. 2021;71(10):5056.

Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–7.

Namsolleck P, Thiel R, Lawson P, Holmstrøm K, Rajilic M, Vaughan EE, et al. Molecular methods for the analysis of gut microbiota. Microb Ecol Health Dis. 2004;16(2–3):71–85.

Liu C, Du MX, Abuduaini R, Yu HY, Li DH, Wang YJ, et al. Enlightening the taxonomy darkness of human gut microbiomes with a cultured biobank. Microbiome. 2021;9(1):119.

Rajilić-Stojanović M, Dimitrijević S, Golić N. Lactic acid bacteria in the gut. In: Lactic acid Bacteria. CRC Press; 2019. p. 383–408.

Tian L, Wang XW, Wu AK, Fan Y, Friedman J, Dahlin A, et al. Deciphering functional redundancy in the human microbiome. Nat Commun. 2020;11(1):6217.

Dai Z, Wu Z, Hang S, Zhu W, Wu G. Amino acid metabolism in intestinal bacteria and its potential implications for mammalian reproduction. MHR Basic Sci Reprod Med. 2015;21(5):389–409.

Stewart BW, De Smet S, Corpet D, Meurillon M, Caderni G, Rohrmann G, et al. Carcinogenicity of consumption of red and processed meat. Lancet Oncol. 2015;16(16):1599–600.

Guyton KZ, Rusyn I, Chiu WA, Corpet DE, van den Berg M, Ross MK, et al. Application of the key characteristics of carcinogens in cancer hazard identification. Carcinogenesis. 2018;39(4):614–22.

Chan DSM, Lau R, Aune D, Vieira R, Greenwood DC, Kampman E, et al. Red and processed meat and colorectal cancer incidence: meta-analysis of prospective studies. PLoS One. 2011;6(6):e20456.

Chazelas E, Pierre F, Druesne-Pecollo N, Esseddik Y, de Edelenyi F, Agaesse C, et al. Nitrites and nitrates from food additives and natural sources and cancer risk: results from the NutriNet-Santé cohort. Int J Epidemiol. 2022;51(4):1106–19.

Xie Y, Geng Y, Yao J, Ji J, Chen F, Xiao J, et al. N-nitrosamines in processed meats: Exposure, formation and mitigation strategies. J Agric Food Res. 2023;13:100645.

Joosen AMCP, Kuhnle GGC, Aspinall SM, Barrow TM, Lecommandeur E, Azqueta A, et al. Effect of processed and red meat on endogenous nitrosation and DNA damage. Carcinogenesis. 2009;30(8):1402–7.

Russell WR, Gratz SW, Duncan SH, Holtrop G, Ince J, Scobbie L, et al. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am J Clin Nutr. 2011;93(5):1062–72.

Massey RC, Key PE, Mallett AK, Rowland IR. An investigation of the endogenous formation of apparent total N-nitroso compounds in conventional microflora and germ-free rats. Food Chem Toxicol. 1988;26(7):595–600.

Suzuki K, Mitsuoka T. N-nitrosamine formation by intestinal bacteria. IARC Sci Publ. 1984;(57):275–81.

Wirbel J, Pyl PT, Kartal E, Zych K, Kashani A, Milanese A, et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med. 2019;25(4):679–89.

Ternes D, Tsenkova M, Pozdeev VI, Meyers M, Koncina E, Atatri S, et al. Author Correction: The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat Metab. 2023;5(9):1638.

Carmody RN, Turnbaugh PJ, others. Host-microbial interactions in the metabolism of therapeutic and diet-derived xenobiotics. J Clin Invest. 2014;124(10):4173–81.

Rampelli S, Soverini M, D’Amico F, Barone M, Tavella T, Monti D, et al. Shotgun metagenomics of gut microbiota in humans with up to extreme longevity and the increasing role of xenobiotic degradation. Msystems. 2020;5(2):10–1128.

Attene-Ramos MS, Wagner ED, Plewa MJ, Gaskins HR. Evidence that hydrogen sulfide is a genotoxic agent. Mol Cancer Res. 2006;4(1):9–14.

Cai WJ, Wang MJ, Ju LH, Wang C, Zhu YC. Hydrogen sulfide induces human colon cancer cell proliferation: role of Akt, ERK and p21. Cell Biol Int. 2010;34(6):565–72.

Magee EA, Richardson CJ, Hughes R, Cummings JH. Contribution of dietary protein to sulfide production in the large intestine: an in vitro and a controlled feeding study in humans. Am J Clin Nutr. 2000;72(6):1488–94.

Wolf PG, Cowley ES, Breister A, Matatov S, Lucio L, Polak P, et al. Diversity and distribution of sulfur metabolic genes in the human gut microbiome and their association with colorectal cancer. Microbiome. 2022;10(1):64.

Du P, Tseng Y, Liu P, Zhang H, Huang G, Chen J, et al. Role of exhaled hydrogen sulfide in the diagnosis of colorectal cancer. BMJ Open Gastroenterol. 2024;11(1):e001229.

Szabo C, Coletta C, Chao C, Módis K, Szczesny B, Papapetropoulos A, et al. Tumorderived hydrogen sulfide, produced by cystathionine$β$-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc Natl Acad Sci. 2013;110(30):12474–9.

Al Hinai EA, Kullamethee P, Rowland IR, Swann J, Walton GE, Commane DM. Modelling the role of microbial p-cresol in colorectal genotoxicity. Gut Microbes. 2019;10(3):398–411.

Pinto J, Carapito Â, Amaro F, Lima AR, Carvalho-Maia C, Martins MC, et al. Discovery of volatile biomarkers for bladder cancer detection and staging through urine metabolomics. Metabolites. 2021;11(4):199.

Andriamihaja M, Lan A, Beaumont M, Audebert M, Wong X, Yamada K, et al. The deleterious metabolic and genotoxic effects of the bacterial metabolite p-cresol on colonic epithelial cells. Free Radic Biol Med. 2015;85:219–27.

Saito Y, Sato T, Nomoto K, Tsuji H. Identification of phenol-and p-cresol-producing intestinal bacteria by using media supplemented with tyrosine and its metabolites. FEMS Microbiol Ecol. 2018;94(9):fiy125.

Brix LA, Barnett AC, Duggleby RG, Leggett B, McManus ME. Analysis of the substrate specificity of human sulfotransferases SULT1A1 and SULT1A3: site-directed mutagenesis and kinetic studies. Biochemistry. 1999;38(32):10474–9.

Gryp T, Vanholder R, Vaneechoutte M, Glorieux G. p-Cresyl sulfate. Toxins (Basel). 2017;9(2):52.

Patel KP, Luo FJG, Plummer NS, Hostetter TH, Meyer TW. The production of pcresol sulfate and indoxyl sulfate in vegetarians versus omnivores. Clin J Am Soc Nephrol. 2012;7(6):982–8.

Bays HE, Taub PR, Epstein E, Michos ED, Ferraro RA, Bailey AL, et al. Ten things to know about ten cardiovascular disease risk factors. Am J Prev Cardiol. 2021;5:100149.

Würtz P, Havulinna AS, Soininen P, Tynkkynen T, Prieto-Merino D, Tillin T, et al. Metabolite profiling and cardiovascular event risk: a prospective study of 3 population-based cohorts. Circulation. 2015;131(9):774–85.

Witkowski M, Weeks TL, Hazen SL. Gut microbiota and cardiovascular disease. Circ Res. 2020;127(4):553–70.

Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, et al. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell. 2020;180(5):862–77.

Nemet I, Li XS, Haghikia A, Li L, Wilcox J, Romano KA, et al. Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality. Eur Heart J. 2023;44(32):3085–96.

Chiu CA, Lu LF, Yu TH, Hung WC, Chung FM, Tsai IT, et al. Increased levels of total P-Cresylsulphate and indoxyl sulphate are associated with coronary artery disease in patients with diabetic nephropathy. Rev Diabet Stud RDS. 2010;7(4):275.

Bredt DS. Endogenous nitric oxide synthesis: biological functions and pathophysiology. Free Radic Res. 1999;31(6):577–96.

Gheibi S, Ghasemi A. Insulin secretion: The nitric oxide controversy. EXCLI J. 2020;19:1227.

Bryan NS, Tribble G, Angelov N. Oral microbiome and nitric oxide: the missing link in the management of blood pressure. Curr Hypertens Rep. 2017;19:1–8.

der Heiden C, Wadman SK, De Bree PK, Wauters EAK. Increased urinary imidazolepropionic acid, n-acetylhistamine and other imidazole compounds in patients with intestinal disorders. Clin Chim Acta. 1972;39(1):201–14.

Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175(4):947–61.

Molinaro A, Bel Lassen P, Henricsson M, Wu H, Adriouch S, Belda E, et al. Imidazole propionate is increased in diabetes and associated with dietary patterns and altered microbial ecology. Nat Commun. 2020;11(1):5881.

van Son J, Serlie MJ, Ståhlman M, Bäckhed F, Nieuwdorp M, Aron-Wisnewsky J. Plasma imidazole propionate is positively correlated with blood pressure in overweight and obese humans. Nutrients. 2021;13(8):2706.

Molinaro A, Nemet I, Bel Lassen P, Chakaroun R, Nielsen T, Aron-Wisnewsky J, et al. Microbially produced imidazole propionate is associated with heart failure and mortality. Heart Fail. 2023;11(7):810–21.

Rios-Covian D, Gonzalez S, Nogacka AM, Arboleya S, Salazar N, Gueimonde M, et al. An overview on fecal branched short-chain fatty acids along human life and as related with body mass index: associated dietary and anthropometric factors. Front Microbiol. 2020;11:513909.

Macfarlane GT, Gibson GR, Beatty E, Cummings JH. Estimation of short-chain fatty acid production from protein by human intestinal bacteria based on branched-chain fatty acid measurements. FEMS Microbiol Ecol. 1992;10(2):81–8.

Alexeev EE, Lanis JM, Kao DJ, Campbell EL, Kelly CJ, Battista KD, et al. Microbiota-derived indole metabolites promote human and murine intestinal homeostasis through regulation of interleukin-10 receptor. Am J Pathol. 2018;188(5):1183–94.

Cason CA, Dolan KT, Sharma G, Tao M, Kulkarni R, Helenowski IB, et al. Plasma microbiome-modulated indole-and phenyl-derived metabolites associate with advanced atherosclerosis and postoperative outcomes. J Vasc Surg. 2018;68(5):1552–62.

Zheng JS, Steur M, Imamura F, Freisling H, Johnson L, van der Schouw YT, et al. Dietary intake of plant-and animal-derived protein and incident cardiovascular diseases: the pan-European EPIC-CVD case--cohort study. Am J Clin Nutr. 2024;119(5):1164–74.

Krga I. Therapeutics and microbiota. Microbiota Heal Dis. 2022;4:762–74.

Bačić A, Gavrilović J, Rajilić-Stojanović M. Polyphenols as a new class of prebiotics for gut microbiota manipulation. Arch Pharm. 2023;73(6):535–53.

Thomas AM, Manghi P, Asnicar F, Pasolli E, Armanini F, Zolfo M, et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat Med. 2019;25(4):667–78.

Qi J, You T, Li J, Pan T, Xiang L, Han Y, et al. Circulating trimethylamine N-oxide and the risk of cardiovascular diseases: a systematic review and meta-analysis of 11 prospective cohort studies. J Cell Mol Med. 2018;22(1):185–94.

Wilcox J, Skye SM, Graham B, Zabell A, Li XS, Li L, et al. Dietary choline supplements, but not eggs, raise fasting TMAO levels in participants with normal renal function: a randomized clinical trial. Am J Med. 2021;134(9):1160–9.

Objavljeno
2024/06/28
Rubrika
Pregledni (Revijalni) rad