The microbiota and chronic kidney diseases: a double-edged sword

Raphael Jose Ferreira Felizardo, Angela Castoldi, Vinicius Andrade-Oliveira, Niels Olsen Saraiva Câmara, Raphael Jose Ferreira Felizardo, Angela Castoldi, Vinicius Andrade-Oliveira, Niels Olsen Saraiva Câmara

Abstract

Recent findings regarding the influence of the microbiota in many inflammatory processes have provided a new way to treat diseases. Now, one may hypothesize that the origin of a plethora of diseases is related to the health of the gut microbiota and its delicate, although complex, interface with the epithelial and immune systems. The 'westernization' of diets, for example, is associated with alterations in the gut microbiota. Such alterations have been found to correlate directly with the increased incidence of diabetes and hypertension, the main causes of chronic kidney diseases (CKDs), which, in turn, have a high estimated prevalence. Indeed, data have arisen showing that the progression of kidney diseases is strictly related to the composition of the microbiota. Alterations in the gut microbiota diversity during CKDs do not only have the potential to exacerbate renal injury but may also contribute to the development of associated comorbidities, such as cardiovascular diseases and insulin resistance. In this review, we discuss how dysbiosis through alterations in the gut barrier and the consequent activation of immune system could intensify the progression of CKD and vice versa, how CKDs can modify the gut microbiota diversity and abundance.

Figures

Figure 1
Figure 1
Dysbiosis and IgA nephropathy. The balance of gut homeostasis is ruled by microbial composition and secretion of specific immunoglobulins, among them IgA. Gut microbial composition imbalance and the high secretion of under glycosylated oligomeric IgA against gut bacteria may trigger the development of IgA nephropathy. LPS, lipopolysaccharide.
Figure 2
Figure 2
Dybiosis and the evolution of kidney damage. In a healthy intestinal microflora, symbiotic bacteria coexist with pathogenic bacteria. The outnumbered presence of symbionts is supposed to maintain gut homeostasis by degrading resistant carbohydrates and proteins that escape from gastric metabolism. An imbalance of the gut biochemical milieu either due to the changes in dietary habits, in which the supply of non-metabolized proteins that reach the intestine is higher than the supply of carbohydrates, or due to the biochemical alterations resulting from reduced kidney function, which impairs the excretion of metabolism products such as urea or acid, may reinforce or contribute to dysbiosis. The reduction of mucus production, loss of tight junctions, leakage of the gut barrier and bacterial translocation initiated due to the overproduction of nitrogen products by urease and uricase-producing bacteria generate high levels of indole sulfate and p-cresil, which, along with endotoxins and LPS, cross the lamina propria to reach the kidneys. The activation of TLRs and other pattern recognition receptors by endotoxins as well as the binding of AhRs by indoxyl sulfate in podocytes are involved in podocyte death and, consequently, the loss of renal selectivity, increased permeability to high-molecular-weight proteins, such as albumin, and renal failure. The damage is propagated to other cells on subjacent glomeruli and progressively contributes to decreased renal clearance. Higher levels of nitrogen products in the blood achieve the intestinal lumen and reinforce, in a vicious cycle, dysbiosis. AhR, aryl hydrocarbon receptor; GBM, glomerular basement membrane. LPS, lipopolysaccharide; TLR, Toll-like receptor; Treg, regulatory T cell.

References

    1. Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B et al. Chronic kidney disease: global dimension and perspectives. Lancet 2013; 382: 260–272.
    1. Cordain L, Eaton SB, Sebastian A, Mann N, Lindeberg S, Watkins BA et al. Origins and evolution of the Western diet: health implications for the 21st century. Am J Clin Nutr 2005; 81: 341–354.
    1. Pereira MA, Kartashov AI, Ebbeling CB, Van Horn L, Slattery ML, Jacobs DR Jr et al. Fast-food habits, weight gain, and insulin resistance (the CARDIA study): 15-year prospective analysis. Lancet 2005; 365: 36–42.
    1. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M et al. Diversity of the human intestinal microbial flora. Science 2005; 308: 1635–1638.
    1. Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G et al. The gut microbiota and host health: a new clinical frontier. Gut 2016; 65: 330–339.
    1. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013; 54: 2325–2340.
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014; 505: 559–563.
    1. Goldszmid RS, Trinchieri G. The price of immunity. Nat Immunol 2012; 13: 932–938.
    1. Chung WS, Walker AW, Louis P, Parkhill J, Vermeiren J, Bosscher D et al. Modulation of the human gut microbiota by dietary fibres occurs at the species level. BMC Biol 2016; 14: 3.
    1. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS et al. Metagenomic analysis of the human distal gut microbiome. Science 2006; 312: 1355–1359.
    1. Macfarlane GT, Macfarlane S. Fermentation in the human large intestine: its physiologic consequences and the potential contribution of prebiotics. J Clin Gastroenterol 2011; 45: S120–S127.
    1. Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and "western-lifestyle" inflammatory diseases. Immunity 2014; 40: 833–842.
    1. Thorburn AN, McKenzie CI, Shen S, Stanley D, Macia L, Mason LJ et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat Commun 2015; 6: 7320.
    1. Evenepoel P, Meijers BK, Bammens BR, Verbeke K. Uremic toxins originating from colonic microbial metabolism. Kidney Int Suppl 2009; 76: S12–S19.
    1. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA 2010; 107: 14691–14696.
    1. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 2014; 20: 159–166.
    1. Berthon BS, Macdonald-Wicks LK, Gibson PG, Wood LG. Investigation of the association between dietary intake, disease severity and airway inflammation in asthma. Respirology 2013; 18: 447–454.
    1. Ou J, Carbonero F, Zoetendal EG, DeLany JP, Wang M, Newton K et al. Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans. Am J Clin Nutr 2013; 98: 111–120.
    1. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med 2015; 7: 307ra152.
    1. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013; 341: 1241214.
    1. Vaziri ND, Wong J, Pahl M, Piceno YM, Yuan J, DeSantis TZ et al. Chronic kidney disease alters intestinal microbial flora. Kidney Int 2013; 83: 308–315.
    1. Vaziri ND, Liu SM, Lau WL, Khazaeli M, Nazertehrani S, Farzaneh SH et al. High amylose resistant starch diet ameliorates oxidative stress, inflammation, and progression of chronic kidney disease. PLoS ONE 2014; 9: e114881.
    1. Richardson AJ, McKain N, Wallace RJ. Ammonia production by human faecal bacteria, and the enumeration, isolation and characterization of bacteria capable of growth on peptides and amino acids. BMC Microbiol 2013; 13: 6.
    1. Kang JY. The gastrointestinal tract in uremia. Dig Dis Sci 1993; 38: 257–268.
    1. Hatch M, Freel RW, Vaziri ND. Intestinal excretion of oxalate in chronic renal failure. J Am Soc Nephrol 1994; 5: 1339–1343.
    1. Siener R, Bangen U, Sidhu H, Hönow R, von Unruh G, Hesse A. The role of Oxalobacter formigenes colonization in calcium oxalate stone disease. Kidney Int 2013; 83: 1144–1149.
    1. Hatch M, Vaziri ND. Enhanced enteric excretion of urate in rats with chronic renal failure. Clin Sci (Lond) 1994; 86: 511–516.
    1. Gower BA, Bergman R, Stefanovski D, Darnell B, Ovalle F, Fisher G et al. Baseline insulin sensitivity affects response to high-amylose maize resistant starch in women: a randomized, controlled trial. Nutr Metab (Lond) 2016; 13: 2.
    1. Lin CH, Chang DM, Wu DJ, Peng HY, Chuang LM. Assessment of blood glucose regulation and safety of resistant starch formula-based diet in healthy normal and subjects with type 2 diabetes. Medicine (Baltimore) 2015; 94: e1332.
    1. Keenan MJ, Martin RJ, Raggio AM, McCutcheon KL, Brown IL, Birkett A et al. High-amylose resistant starch increases hormones and improves structure and function of the gastrointestinal tract: a microarray study. J Nutrigenet Nutrigenomics 2012; 5: 26–44.
    1. Macia L, Tan J, Vieira AT, Leach K, Stanley D, Luong S et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun 2015; 6: 6734.
    1. Valcheva R, Hotte N, Gillevet P, Sikaroodi M, Thiessen A, Madsen KL. Soluble dextrin fibers alter the intestinal microbiota and reduce proinflammatory cytokine secretion in male il-10-deficient mice. J Nutr 2015; 145: 2060–2066.
    1. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013; 341: 569–573.
    1. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013; 504: 451–455.
    1. Chang PV, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci USA 2014; 111: 2247–2252.
    1. Braga TT, Correa-Costa M, Guise YF, Castoldi A, de Oliveira CD, Hyane MI et al. MyD88 signaling pathway is involved in renal fibrosis by favoring a TH2 immune response and activating alternative M2 macrophages. Mol Med 2012; 18: 1231–1239.
    1. Correa-Costa M, Braga TT, Felizardo RJ, Andrade-Oliveira V, Perez KR, Cuccovia IM et al. Macrophage trafficking as key mediator of adenine-induced kidney injury. Mediators Inflamm 2014; 2014: 291024.
    1. Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A, Felizardo RJ, de Almeida DC et al. Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. J Am Soc Nephrol 2015; 26: 1877–1888.
    1. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA 2013; 110: 4410–4415.
    1. Van Beneden K, Geers C, Pauwels M, Mannaerts I, Verbeelen D, van Grunsven LA et al. Valproic acid attenuates proteinuria and kidney injury. J Am Soc Nephrol 2011; 22: 1863–1875.
    1. Ichii O, Otsuka-Kanazawa S, Nakamura T, Ueno M, Kon Y, Chen W et al. Podocyte injury caused by indoxyl sulfate, a uremic toxin and aryl-hydrocarbon receptor ligand. PLoS ONE 2014; 9: e108448.
    1. Vaziri ND, Goshtasbi N, Yuan J, Jellbauer S, Moradi H, Raffatellu M et al. Uremic plasma impairs barrier function and depletes the tight junction protein constituents of intestinal epithelium. Am J Nephrol 2012; 36: 438–443.
    1. Bossola M, Sanguinetti M, Scribano D, Zuppi C, Giungi S, Luciani G et al. Circulating bacterial-derived DNA fragments and markers of inflammation in chronic hemodialysis patients. Clin J Am Soc Nephrol 2009; 4: 379–385.
    1. Wang F, Jiang H, Shi K, Ren Y, Zhang P, Cheng S et al. Gut bacterial translocation is associated with microinflammation in end-stage renal disease patients. Nephrology (Carlton) 2012; 17: 733–738.
    1. Leemans JC, Kors L, Anders HJ, Florquin S. Pattern recognition receptors and the inflammasome in kidney disease. Nat Rev Nephrol 2014; 10: 398–414.
    1. Castoldi A, Braga TT, Correa-Costa M, Aguiar CF, Bassi ÊJ, Correa-Silva R et al. TLR2, TLR4 and the MYD88 signaling pathway are crucial for neutrophil migration in acute kidney injury induced by sepsis. PLoS ONE 2012; 7: e37584.
    1. Correa-Costa M, Braga TT, Semedo P, Hayashida CY, Bechara LR, Elias RM et al. Pivotal role of Toll-like receptors 2 and 4, its adaptor molecule MyD88, and inflammasome complex in experimental tubule-interstitial nephritis. PLoS ONE 2011; 6: e29004.
    1. Wolfs TG, Buurman WA, van Schadewijk A, de Vries B, Daemen MA, Hiemstra PS et al. In vivo expression of Toll-like receptor 2 and 4 by renal epithelial cells: IFN-gamma and TNF-alpha mediated up-regulation during inflammation. J Immunol 2002; 168: 1286–1293.
    1. Du P, Fan B, Han H, Zhen J, Shang J, Wang X et al. NOD2 promotes renal injury by exacerbating inflammation and podocyte insulin resistance in diabetic nephropathy. Kidney Int 2013; 84: 265–276.
    1. Granata S, Masola V, Zoratti E, Scupoli MT, Baruzzi A, Messa M et al. NLRP3 inflammasome activation in dialyzed chronic kidney disease patients. PLoS ONE 2015; 10: e0122272.
    1. Seo SU, Kamada N, Muñoz-Planillo R, Kim YG, Kim D, Koizumi Y et al. Distinct commensals induce interleukin-1β via NLRP3 inflammasome in inflammatory monocytes to promote intestinal inflammation in response to injury. Immunity 2015; 42: 744–755.
    1. Dalal SR, Chang EB. The microbial basis of inflammatory bowel diseases. J Clin Invest 2014; 124: 4190–4196.
    1. De Angelis M, Montemurno E, Piccolo M, Vannini L, Lauriero G, Maranzano V et al. Microbiota and metabolome associated with immunoglobulin A nephropathy (IgAN). PLoS ONE 2014; 9: e99006.
    1. Kamata T, Nogaki F, Fagarasan S, Sakiyama T, Kobayashi I, Miyawaki S et al. Increased frequency of surface IgA-positive plasma cells in the intestinal lamina propria and decreased IgA excretion in hyper IgA (HIGA) mice, a murine model of IgA nephropathy with hyperserum IgA. J Immunol 2000; 165: 1387–1394.
    1. Papista C, Lechner S, Ben Mkaddem S, LeStang MB, Abbad L, Bex-Coudrat J et al. Gluten exacerbates IgA nephropathy in humanized mice through gliadin-CD89 interaction. Kidney Int 2015; 88: 276–285.
    1. Lechner SM, Papista C, Chemouny JM, Berthelot L, Monteiro RC. Role of IgA receptors in the pathogenesis of IgA nephropathy. J Nephrol 2016; 29: 5–11.
    1. Coppo R, Amore A, Hogg R, Emancipator S. Idiopathic nephropathy with IgA deposits. Pediatr Nephrol 2000; 15: 139–150.
    1. Almroth G, Axelsson T, Müssener E, Grodzinsky E, Midhagen G, Olcén P. Increased prevalence of anti-gliadin IgA-antibodies with aberrant duodenal histopathological findings in patients with IgA-nephropathy and related disorders. Ups J Med Sci 2006; 111: 339–352.
    1. McCarthy DD, Kujawa J, Wilson C, Papandile A, Poreci U, Porfilio EA et al. Mice overexpressing BAFF develop a commensal flora-dependent, IgA-associated nephropathy. J Clin Invest 2011; 121: 3991–4002.
    1. Filiopoulos V, Trompouki S, Hadjiyannakos D, Paraskevakou H, Kamperoglou D, Vlassopoulos D. IgA nephropathy in association with Crohn's disease: a case report and brief review of the literature. Ren Fail 2010; 32: 523–527.
    1. Krishnamurthy VM, Wei G, Baird BC, Murtaugh M, Chonchol MB, Raphael KL et al. High dietary fiber intake is associated with decreased inflammation and all-cause mortality in patients with chronic kidney disease. Kidney Int 2012; 81: 300–306.
    1. Daniel CR, Park Y, Chow WH, Graubard BI, Hollenbeck AR, Sinha R. Intake of fiber and fiber-rich plant foods is associated with a lower risk of renal cell carcinoma in a large US cohort. Am J Clin Nutr 2013; 97: 1036–1043.
    1. Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol 2014; 39: 230–237.
    1. Barreto FC, Barreto DV, Liabeuf S, Meert N, Glorieux G, Temmar M et al. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol 2009; 4: 1551–1558.
    1. Dou L, Jourde-Chiche N, Faure V, Cerini C, Berland Y, Dignat-George F et al. The uremic solute indoxyl sulfate induces oxidative stress in endothelial cells. J Thromb Haemost 2007; 5: 1302–1308.
    1. Neirynck N, Vanholder R, Schepers E, Eloot S, Pletinck A, Glorieux G. An update on uremic toxins. Int Urol Nephrol 2013; 45: 139–150.
    1. Niwa T, Shimizu H. Indoxyl sulfate induces nephrovascular senescence. J Ren Nutr 2012; 22: 102–106.
    1. Niwa T, Ise M, Miyazaki T. Progression of glomerular sclerosis in experimental uremic rats by administration of indole, a precursor of indoxyl sulfate. Am J Nephrol 1994; 14: 207–212.
    1. Enomoto A, Takeda M, Tojo A, Sekine T, Cha SH, Khamdang S et al. Role of organic anion transporters in the tubular transport of indoxyl sulfate and the induction of its nephrotoxicity. J Am Soc Nephrol 2002; 13: 1711–1720.
    1. Grahammer F, Schell C, Huber TB. The podocyte slit diaphragm—from a thin grey line to a complex signalling hub. Nat Rev Nephrol 2013; 9: 587–598.
    1. Poesen R, Windey K, Neven E, Kuypers D, De Preter V, Augustijns P et al. The Influence of CKD on Colonic Microbial Metabolism. J Am Soc Nephrol 2015; 27: 1389–1399.
    1. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol 2015; 8: 80–93.
    1. Rossi M, Johnson DW, Xu H, Carrero JJ, Pascoe E, French C et al. Dietary protein-fiber ratio associates with circulating levels of indoxyl sulfate and p-cresyl sulfate in chronic kidney disease patients. Nutr Metab Cardiovasc Dis 2015; 25: 860–865.
    1. Meijers BK, De Preter V, Verbeke K, Vanrenterghem Y, Evenepoel P. p-Cresyl sulfate serum concentrations in haemodialysis patients are reduced by the prebiotic oligofructose-enriched inulin. Nephrol Dial Transplant 2010; 25: 219–224.
    1. Rossi M, Johnson DW, Morrison M, Pascoe EM, Coombes JS, Forbes JM et al. Synbiotics easing renal failure by improving gut microbiology (SYNERGY): a randomized trial. Clin J Am Soc Nephrol 2016; 11: 223–231.
    1. Wang H, Zhang W, Zuo L, Zhu W, Wang B, Li Q et al. Bifidobacteria may be beneficial to intestinal microbiota and reduction of bacterial translocation in mice following ischaemia and reperfusion injury. Br J Nutr 2013; 109: 1990–1998.
    1. Ranganathan N, Ranganathan P, Friedman EA, Joseph A, Delano B, Goldfarb DS et al. Pilot study of probiotic dietary supplementation for promoting healthy kidney function in patients with chronic kidney disease. Adv Ther 2010; 27: 634–647.

Source: PubMed

3
Se inscrever