Next Article in Journal
A Review on Mycotoxins and Microfungi in Spices in the Light of the Last Five Years
Next Article in Special Issue
Thrombolome and Its Emerging Role in Chronic Kidney Diseases
Previous Article in Journal
Exposure to Aerosolized Algal Toxins in South Florida Increases Short- and Long-Term Health Risk in Drosophila Model of Aging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Links between Microbiome and Uremic Toxins in Acute Kidney Injury: Beyond Gut Feeling—A Systematic Review

by
Alicja Rydzewska-Rosołowska
1,*,
Natalia Sroka
1,
Katarzyna Kakareko
1,
Mariusz Rosołowski
2,
Edyta Zbroch
1 and
Tomasz Hryszko
1
1
2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
2
Department of Gastroenterology and Internal Medicine, Medical University of Białystok, 15-276 Białystok, Poland
*
Author to whom correspondence should be addressed.
Submission received: 30 October 2020 / Revised: 7 December 2020 / Accepted: 9 December 2020 / Published: 11 December 2020
(This article belongs to the Special Issue Study on the Uremic Toxin Targeting Mechanism)

Abstract

:
The last years have brought an abundance of data on the existence of a gut-kidney axis and the importance of microbiome in kidney injury. Data on kidney-gut crosstalk suggest the possibility that microbiota alter renal inflammation; we therefore aimed to answer questions about the role of microbiome and gut-derived toxins in acute kidney injury. PubMed and Cochrane Library were searched from inception to October 10, 2020 for relevant studies with an additional search performed on ClinicalTrials.gov. We identified 33 eligible articles and one ongoing trial (21 original studies and 12 reviews/commentaries), which were included in this systematic review. Experimental studies prove the existence of a kidney-gut axis, focusing on the role of gut-derived uremic toxins and providing concepts that modification of the microbiota composition may result in better AKI outcomes. Small interventional studies in animal models and in humans show promising results, therefore, microbiome-targeted therapy for AKI treatment might be a promising possibility.
Key Contribution: The article is a systematic review of available data on the kidney-gut axis and the role of gut-derived uremic toxins in the course of acute kidney injury.

1. Introduction

The term microbiota is used to describe a community of 100 trillion microorganisms (more than 1000 species, of mostly bacteria, but also viruses, fungi, and protozoa) that are present in the gastrointestinal tract. The term microbiome on the other hand, refers to the collective genomes of the above-mentioned organisms. The microbiome encodes over 3 million genes (as compared to 30,000 genes present in the human genome) characterized now by the Human Microbiome Project [1], which produce thousands of metabolites. Unsurprisingly, those metabolites play an important role both in human biology and disease development [2].
In healthy subjects, the phyla Bacteroidetes and Firmicutes dominate, composing more than 90% of all species, although density and species composition varies alongside the digestive tract [3]. The functional diversity of microbiota is key to normal biology.
Kidney failure directly or indirectly modifies microbial composition in the digestive tract. Edema and ischemia of the intestinal wall complicating kidney diseases lead to increased intestinal permeability, or so-called “leaky gut”, which further activates the immune system and results in systemic inflammation.
The relationship is bidirectional, as dysbiosis (changes in structure and composition of the microbiome) is implicated in many kidney diseases. The existence of a gut-kidney axis has been extensively proved. Communication between those two organs occurs not only directly, but also through the aforementioned metabolites. We have long known that many uremic toxins, such as ammonia, indole sulfate, p-cresyl sulfate, indole-3 acetic acid, trimethylamine N-oxide (TMAO) are gut-derived. Short chain fatty acids (SCFAs)–anaerobic fermentation end products of complex carbohydrates, on the other hand, are insufficiently generated due to dysbiosis. Most research focuses on chronic kidney disease (CKD) and the unmistakable role these toxins play in cardiovascular complications, mortality, or disease progression [4]. Less is known about the gut-kidney crosstalk present in acute kidney injury (AKI).

2. Objective

This systematic review aims to answer questions about the role of microbiome and gut-derived toxins in acute kidney injury. We analyzed both experimental and interventional data, animal and human studies, original articles and reviews/opinions aiming to gather full knowledge on the subject in question.

3. Results

Out of 1014 identified records, 638 were screened, 36 retrieved and assessed for eligibility and 33 finally included in the review (21 original studies and 12 reviews/commentaries). The entire selection process is illustrated in Figure 1. The basic characteristics of the original articles on the researched subject are summarized in Table 1 and Table 2. We have identified 16 experimental studies and five human studies (three prospective observational and two retrospective). We could therefore not assess the risk of bias and applicability concerns. ClinicalTrials.gov identified 226 studies with 36 duplicates, and after screening, one study, pertaining to the subject of the review remained.

4. Discussion

Acute kidney injury is usually multifactorial and involves hemodynamic, biochemical, inflammatory, and immunological mechanisms. The first studies on the influence of microbiome on AKI stemmed from the “hygiene hypothesis”, which states that the lack of exposure to normal microbiota induces less immune deviation and reduces immune regulation [26]. Jang and coworkers studied the effects of ischemia-reperfusion injury (IRI) on germ-free mice and found that compared to controls, the severity of injury (measured with creatinine and histologically determining the extent of tubular injury) was higher. After addition of bacteria to the diet, renal injury was equivalent to that of control mice [5].

4.1. The Gut-Kidney-Axis

There is a multitude of evidence that CKD alters microbiome composition [27]. Experimental animal evidence suggests that such is also the case with acute kidney injury. The relationship is bidirectional—AKI not only causes dysbiosis, microbiota composition also determines the severity of kidney injury [6,9,10]. In a murine model, hypoxia altered the ratio of aerobic/anaerobic population, increased the quantity of strict anaerobes and total lactic acid bacteria [6]. In another experimental setting of ischemia reperfusion injury, the balance between Rothia and Streptococcus genera was associated with creatinine levels [10].
Gut microbiota affect not only the levels of certain uremic toxins but both adaptive and innate immune responses. The numbers of natural killer T cells were higher in germ-free mice, AKI induction resulted in a greater number of CD8 cells [5]. Th17 cells are also crucial in kidney injury, as they produce Il-17 and directly induce renal inflammation by neutrophil activation and macrophage-mediated injury. Emerging evidence shows that the immunomodulatory effects of the intestinal microbiota include Th17 induction [28].
Another key link between the gut and the kidney is immunoglobulin A (IgA). It limits bacterial association with the epithelium [29] and prevents bacterial penetration of host tissue [30]. The exact mechanisms by which it works still remain unclear. Clues from research on IgA nephropathy also point to existence of an IgA axis between the mucosa, the bone marrow, and the kidney. In that specific disease, genome-wide association studies have identified risk loci in genes involved in the maintenance of the intestinal epithelial barrier and response to mucosal pathogens. The genetic risk also strongly correlates with microbiota variation, particularly helminth diversity [31]. The role of IgA in acute kidney injury is still unknown.
Drugs used for kidney disease treatment also affect the gut-kidney axis. One of the examples is oral iron, which increases gut microbial protein fermentation and in turn causes an increase in fecal and plasma uremic toxin levels. Increase in gut transit time may further increase the levels of many uremic toxins (such as phenols and indoles) [32,33]. The effect of many drugs used for AKI treatment on microbiome remains largely unknown.
As all intestinal barriers, biological, physical, and immune barriers might become damaged during kidney injury (the aforementioned “leaky gut hypothesis”), as levels of other toxins, mainly inflammatory, rise. Circulating lipopolysaccharide and endotoxin levels are higher during AKI and stimulate intrarenal inflammatory response [8].
A schematic review of the gut-kidney axis in AKI is shown in Figure 2.

4.2. Uremic Toxins

Healthy kidneys excrete a multitude of substances. In patients with CKD retention, these substances occur. These substances, when they contribute to the development of uremia, are called uremic toxins. They can be divided into three categories: small water-soluble, non-protein-bound molecules (such as urea, guanidines, oxalate, TMAO, phosphorus), small, lipid-soluble or protein-bound molecules (such as p-cresol sulfate, indoles, homocysteine), and the so-called middle molecules such as beta-2 microglobulin. It is well-known that some of these toxins are produced by the microbiota. Dysbiosis leads to excessive secretion of gut-derived uremic toxins, which in turn may further damage renal tubular cells [34].
Experimental evidence shows that selective modification of the microbiome might change the uremic toxin profile, e.g., in mice, deleting the tryptophanase gene from Bacteroidetes present in the digestive tract resulted in nearly undetectable indole sulfate levels [35]. Both indole sulfate and p-cresyl sulfate are well-researched toxins and their levels correlate with cardiovascular events and mortality in dialysis patients [36,37]. The levels of those toxins rise in acute kidney injury and correlate with RIFLE classification of AKI severity [15]. In a prospective cohort study, serum indoxyl sulfate levels were also associated with mortality in hospital-acquired AKI [14]. In a murine model, suppression of indole and p-cresyl production by Lactobacillus salivarius protected against cisplatin-induced kidney injury [23].
Trimethylamine-N-oxide is a gut-derived metabolite of phosphatidylcholine and a known uremic toxin. It directly enhances atherogenesis. There is a proven correlation between elevated TMAO levels and major adverse cardiovascular events (death, myocardial infarction, or stroke) [38]. CKD patients (including hemodialysis) with elevated TMAO levels have lower long-term survival [39,40]. Studies in AKI on TMAO are still pending.
Homocysteine (Hcy), a transmethylation product of metabolic conversion of methionine to cysteine, is an extensively studied gut-derived uremic toxin. Hyperhomocysteinemia is implicated in the progression of chronic kidney disease, and many cardiovascular complications [41,42]. In an experimental murine model, it induced more severe cisplatin induced-AKI than in mice with normal Hcy levels [7]. Unfortunately, secondary homocysteine lowering in a large randomized clinical trial did not improve survival or reduce the incidence of vascular disease in patients with advanced chronic kidney disease or end-stage kidney disease [43].
Acylcarnitines, metabolites of carnitine, are inversely correlated with blood pressure and cholesterol levels in hemodialysis patients [44]. In another murine model an increase in the levels of acylcarnitines was detected after ischemia-reperfusion injury of the kidney together with associations of certain bacterial species and metabolite levels (Rothia and Staphylococcus abundance positively correlated with severity of AKI) [10]. The exact mechanism is still unknown but suggests potential targets for future therapeutic interventions.
Hippurate on the other hand is reported by some to be a metabolomic marker of gut microbiome diversity [45]. In a 1994 study of 35 kidney transplant recipients, hippuric acid levels were significantly higher in patients with acute allograft rejection. It was hypothesized that this is due to reduced excretion; blood levels fell after successful antirejection treatment and microbiota composition was not assessed [12] but it might be another suggestion warranting further research.

4.3. Potential Interventions

Simple nutritional interventions, that modify gut microbiota, might lower uremic toxin production. Oral adsorbents, prebiotics, probiotics, synbiotics, eubiotics or fecal microbiota transplantation (FMT) are possible therapeutic options (Figure 3).
One strategy to decrease gut-derived uremic toxins is adsorbent use. Oral uremic toxin adsorbents are present on the market and have been proven to work. AST-120 (marketed as Kremezin®) binds many low-molecular-weight compounds with superior adsorption of p-cresol sulfate and indoles. In some human phase II and phase III studies it slowed CKD progression, but unequivocal results have not been demonstrated [46,47]. In a murine model, treatment with AST-120 reduced indole levels and decreased renal expression of mRNAs of injury-related markers [19].
Prebiotics are defined as “selectively fermented ingredients that result in specific changes in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefit(s) upon host health” [48]. The role of different prebiotics in CKD has been studied with promising results; in a prospective crossover single-blind study gum arabic fiber combined with low-protein diet patients had lower serum urea nitrogen levels [49]. We have not found any studies on the use of prebiotics in AKI; a trial NCT03877081 on the effect of probiotics and prebiotics on renal function in septic acute kidney injury patients is registered on ClinicalTrials.gov [50]. On the other hand, anaerobic fermentation of prebiotics results in the production of short chain fatty acids: acetate, butyrate and propionate. SCFAs are recognized as potential mediators modifying intestinal immune function (they modulate inflammatory gene expression, chemotaxis, cell differentiation, proliferation and apoptosis and also improve the course of many inflammatory diseases) [51]. Many experimental studies in mice have shown that SCFAs (especially acetate and butyrate) administration improves outcomes of acute kidney injury [16,17,18,22]. The potential mechanisms need more elucidation, but as a decrease in apoptosis and increase in autophagy together with an increase in mitochondrial DNA content have been shown [16,18], it might be energy conservation and improvement of mitochondrial energetics. The role of olfactory receptor 78 (olf78), a type G protein-coupled receptor that is expressed in the juxtaglomerular apparatus and modulates renin secretion in response to SCFAs, is underlined in some studies [52].
Probiotics are “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [53]. Small experimental studies demonstrate that Lactobacillus salivarius or a microbial cocktail may ameliorate acute kidney injury [23,24]. This seems to be in contrast to studies showing that broad-spectrum antibiotics protected against AKI [20], although further elucidation is needed, as we do not know the exact microbiota content after antibiotic treatment (a possibility exists that protective bacteria genres were spared).
Synbiotics are nutritional supplements that combine both prebiotics and probiotics. In small human CKD studies showed lower concentrations of p-cresol but not indoxyl sulfate after synbiotic use, although results are inconclusive [54,55]. Studies in AKI are lacking.
Substances, which induce homeostatic changes in the intestinal microbiota or eubiosis are sometimes called eubiotics. Rifaximin–a poorly absorbed antibiotic lowered TMAO levels in mice [56] but in a randomized control trial in CKD patients failed to lower TMAO, p-cresol sulfate, indoxyl sulfate, kynurenic acid, deoxycholic acid, and inflammatory cytokines but did decrease bacterial richness and diversity [57]. A retrospective study that analyzed patients with hepatic cirrhosis found a reduced incidence rate of AKI and hepatorenal syndrome and a lower need for renal replacement therapy [25].
Fecal microbiota transplant is widely accepted as definitive treatment of Clostridioides difficile infection [58]. A recently completed trial NCT04361097 registered on ClinicalTrials.gov has evaluated fecal microbiota transplantation in CKD, results are as yet unknown [59], another similar trial is currently recruiting [60]. As yet, there is no data on FMT in AKI, but it remains a possible therapeutic strategy.

5. Conclusions

In the beginning of the 20th century, Ilya Ilyich Metchnikoff hypothesized that the human colon functioned only as a waste reservoir, where microbiota produced only “fermentations and putrefaction harmful to the host” [61]. Today the paradigm is completely changing. Increased data provide evidence of existence of a gut-kidney axis in acute kidney injury, mediated in part by uremic toxins. Experimental studies provide concepts that modification of the microbiota composition may result in better AKI outcomes. Small interventional studies in animal models and in humans show promising results, therefore microbiome-targeted therapy for AKI treatment is a promising possibility.

6. Materials and Methods

We undertook a systematic search of MEDLINE through PubMed and COCHRANE LIBRARY database from inception to October 10, 2020.
We searched both with individual keywords and Medical Subject Headings (MeSH) with all subheadings included. Individual keywords used were “acute kidney injury”, “acute renal failure”, “microbiota”, “microbiome”, “uremic toxins”, “gut-derived toxins”, “nephrotoxicity”. MeSH terms included were “acute kidney injury”, “microbiota”, and “uremia middle molecule toxins”. The terms were combined individually using the Boolean operator “AND”. In addition, the references of eligible papers were searched manually for additional records. The results were merged together with duplicates discarded. Remaining articles were screened for relevance (based on their title, abstract, or full text). All articles on the subject of microbiota and gut-derived uremic toxins in the course of acute kidney injury were included due to paucity of data: original studies and reviews/perspectives were both experimental and human. Articles were excluded only if they were clearly related to other subject matters or were not published in English, French, or German. Additionally, ClinicalTrials.gov was searched with simple keywords (kidney, microbiome, microbiota, gut-derived) for currently registered and active studies.

Author Contributions

Conceptualization, A.R.-R., T.H.; methodology, K.K.; software, T.H.; validation, M.R., N.S., K.K. and E.Z.; formal analysis, A.R.-R., N.S., M.R., E.Z.; investigation, M.R.; resources, T.H.; data curation, N.S.; writing—original draft preparation, A.R.-R.; writing—review and editing, N.S., K.K., M.R., E.Z., T.H.; visualization, T.H.; supervision, T.H.; project administration, A.R.-R.; funding acquisition, T.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

In this section you can acknowledge any support given which is not covered by the author contribution or funding sections. This may include administrative and technical support, or donations in kind (e.g., materials used for experiments).

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. NIH Human Microbiome Portfolio Analysis Team. A review of 10 years of human microbiome research activities at the US National Institutes of Health, Fiscal Years 2007–2016. Microbiome 2019, 7, 31. [Google Scholar] [CrossRef] [PubMed]
  2. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ramezani, A.; Raj, D.S. The gut microbiome, kidney disease, and targeted interventions. J. Am. Soc. Nephrol. 2013, 25, 657–670. [Google Scholar] [CrossRef] [Green Version]
  5. Jang, H.R.; Gandolfo, M.T.; Ko, G.J.; Satpute, S.; Racusen, L.; Rabb, H. Early exposure to germs modifies kidney damage and inflammation after experimental ischemia-reperfusion injury. Am. J. Physiol. Physiol. 2009, 297, F1457–F1465. [Google Scholar] [CrossRef] [Green Version]
  6. Nandi, D.K.; Samanta, A.; Patra, A.; Mandal, S.; Roy, S.; Das, K.; Kar, S. Hypoxia: A cause of acute renal failure and alteration of gastrointestinal microbial ecology. Saudi J. Kidney Dis. Transplant. 2018, 29, 879–888. [Google Scholar] [CrossRef]
  7. Long, Y.; Zhen, X.; Zhu, F.; Hu, Z.; Lei, W.; Li, S.; Zha, Y.; Nie, J. Hyperhomocysteinemia Exacerbates Cisplatin-induced Acute Kidney Injury. Int. J. Biol. Sci. 2017, 13, 219–231. [Google Scholar] [CrossRef] [Green Version]
  8. Li, J.; Moturi, K.; Wang, L.; Zhang, K.; Yu, C. Gut derived-endotoxin contributes to inflammation in severe ischemic acute kidney injury. BMC Nephrol. 2019, 20, 16. [Google Scholar] [CrossRef] [Green Version]
  9. Ma, Y.; Peng, X.; Yang, J.; Giovanni, V.; Wang, C. Impacts of functional oligosaccharide on intestinal immune modulation in immunosuppressive mice. Saudi J. Biol. Sci. 2020, 27, 233–241. [Google Scholar] [CrossRef]
  10. Andrianova, N.V.; Popkov, V.A.; Klimenko, N.; Tyakht, A.; Zakharova, E.Y.; Frolova, O.Y.; Zorova, L.D.; Pevzner, I.B.; Zorov, D.B.; Plotnikov, E.Y. Microbiome-Metabolome Signature of Acute Kidney Injury. Metabolites 2020, 10, 142. [Google Scholar] [CrossRef] [Green Version]
  11. Mishima, E.; Ichijo, M.; Kawabe, T.; Kikuchi, K.; Akiyama, Y.; Toyohara, T.; Suzuki, T.; Suzuki, C.; Asao, A.; Ishii, N.; et al. Germ-Free Conditions Modulate Host Purine Metabolism, Exacerbating Adenine-Induced Kidney Damage. Toxins 2020, 12, 547. [Google Scholar] [CrossRef] [PubMed]
  12. Knoflach, A.; Binswanger, U. Serum hippuric acid concentration in renal allograft rejection, ureter obstruction, and tubular necrosis. Transpl. Int. 1994, 7, 17–21. [Google Scholar] [CrossRef] [PubMed]
  13. Carron, C.; De Barros, J.-P.P.; Gaiffe, E.; Deckert, V.; Adda-Rezig, H.; Roubiou, C.; Laheurte, C.; Masson, D.; Simula-Faivre, D.; Louvat, P.; et al. End-Stage Renal Disease-Associated Gut Bacterial Translocation: Evolution and Impact on Chronic Inflammation and Acute Rejection After Renal Transplantation. Front. Immunol. 2019, 10, 1630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Wang, W.; Hao, G.; Pan, Y.; Ma, S.; Yang, T.; Shi, P.; Zhu, Q.; Xie, Y.; Ma, S.; Zhang, Q.; et al. Serum indoxyl sulfate is associated with mortality in hospital-acquired acute kidney injury: A prospective cohort study. BMC Nephrol. 2019, 20, 1–11. [Google Scholar] [CrossRef] [PubMed]
  15. Veldeman, L.; Vanmassenhove, J.; Van Biesen, W.; Massy, Z.A.; Liabeuf, S.; Glorieux, G.; Vanholder, R. Evolution of protein-bound uremic toxins indoxyl sulphate and p-cresyl sulphate in acute kidney injury. Int. Urol. Nephrol. 2019, 51, 293–302. [Google Scholar] [CrossRef] [PubMed]
  16. Machado, R.A.; Constantino, L.D.S.; Tomasi, C.D.; Rojas, H.A.; Vuolo, F.S.; Vitto, M.F.; Cesconetto, P.A.; De Souza, C.T.; Ritter, C.; Dal-Pizzol, F. Sodium butyrate decreases the activation of NF-kappaB reducing inflammation and oxidative damage in the kidney of rats subjected to contrast-induced nephropathy. Nephrol. Dial. Transplant. 2012, 27, 3136–3140. [Google Scholar] [CrossRef] [Green Version]
  17. Sun, X.; Zhang, B.; Hong, X.; Zhang, X.; Kong, X. Histone deacetylase inhibitor, sodium butyrate, attenuates gentamicin-induced nephrotoxicity by increasing prohibitin protein expression in rats. Eur. J. Pharmacol. 2013, 707, 147–154. [Google Scholar] [CrossRef]
  18. Andrade-Oliveira, V.; Amano, M.T.; Correa-Costa, M.; Castoldi, A.; Felizardo, R.J.; De Almeida, D.C.; Bassi, E.J.; Moraes-Vieira, P.M.; Hiyane, M.I.; Rodas, A.C.; et al. Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. J. Am. Soc. Nephrol. 2015, 26, 1877–1888. [Google Scholar] [CrossRef]
  19. Fujii, H.; Yonekura, Y.; Yamashita, Y.; Kono, K.; Nakai, K.; Goto, S.; Sugano, M.; Goto, S.; Fujieda, A.; Ito, Y.; et al. Anti-oxidative effect of AST-120 on kidney injury after myocardial infarction. Br. J. Pharmacol. 2016, 173, 1302–1313. [Google Scholar] [CrossRef] [Green Version]
  20. Emal, D.; Rampanelli, E.; Stroo, I.; Butter, L.M.; Teske, G.J.; Claessen, N.; Stokman, G.; Florquin, S.; Leemans, J.C.; Dessing, M.C. Depletion of Gut Microbiota Protects against Renal Ischemia-Reperfusion Injury. J. Am. Soc. Nephrol. 2016, 28, 1450–1461. [Google Scholar] [CrossRef] [Green Version]
  21. Nakade, Y.; Iwata, Y.; Furuichi, K.; Mita, M.; Hamase, K.; Konno, R.; Miyake, T.; Sakai, N.; Kitajima, S.; Toyama, T.; et al. Gut microbiota–derived D-serine protects against acute kidney injury. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Al-Harbi, N.O.; Nadeem, A.; Ahmad, S.F.; Alotaibi, M.R.; Alasmari, A.F.; Alanazi, W.A.; Al-Harbi, M.M.; El-Sherbeeny, A.M.; Ibrahim, K.E. Short chain fatty acid, acetate ameliorates sepsis-induced acute kidney injury by inhibition of NADPH oxidase signaling in T cells. Int. Immunopharmacol. 2018, 58, 24–31. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, T.; Park, D.; Kim, Y.J.; Lee, I.; Kim, S.; Oh, C.; Kim, J.; Yang, J.; Jo, S.-K. Lactobacillus salivarius BP121 prevents cisplatin-induced acute kidney injury by inhibition of uremic toxins such as indoxyl sulfate and p-cresol sulfate via alleviating dysbiosis. Int. J. Mol. Med. 2020, 45, 1130–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zheng, D.-W.; Pan, P.; Chen, K.-W.; Fan, J.-X.; Li, C.-X.; Cheng, H.; Zhang, X.-Z. An orally delivered microbial cocktail for the removal of nitrogenous metabolic waste in animal models of kidney failure. Nat. Biomed. Eng. 2020, 4, 853–862. [Google Scholar] [CrossRef] [PubMed]
  25. Dong, T.S.; Aronsohn, A.; Reddy, K.G.; Te, H.S. Rifaximin Decreases the Incidence and Severity of Acute Kidney Injury and Hepatorenal Syndrome in Cirrhosis. Dig. Dis. Sci. 2016, 61, 3621–3626. [Google Scholar] [CrossRef] [PubMed]
  26. Martinez, F.D. The coming-of-age of the hygiene hypothesis. Respir. Res. 2001, 2, 129–132. [Google Scholar] [CrossRef] [PubMed]
  27. Vaziri, N.; Wong, J.; Pahl, M.V.; Piceno, Y.M.; Yuan, J.; DeSantis, T.Z.; Ni, Z.; Nguyen, T.-H.; Andersen, G.L. Chronic kidney disease alters intestinal microbial flora. Kidney Int. 2013, 83, 308–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Kitching, A.R.; Holdsworth, S.R. The Emergence of Th17 Cells as Effectors of Renal Injury. J. Am. Soc. Nephrol. 2011, 22, 235–238. [Google Scholar] [CrossRef] [Green Version]
  29. Suzuki, K.; Meek, B.; Doi, Y.; Muramatsu, M.; Chiba, T.; Honjo, T.; Fagarasan, S. Aberrant expansion of segmented filamentous bacteria in IgA-deficient gut. Proc. Natl. Acad. Sci. USA 2004, 101, 1981–1986. [Google Scholar] [CrossRef] [Green Version]
  30. MacPherson, A.J.; Gatto, D.; Sainsbury, E.; Harriman, G.R.; Hengartner, H.; Zinkernagel, R.M. A primitive T cell-independent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science 2000, 288, 2222–2226. [Google Scholar] [CrossRef]
  31. Kiryluk, K.; Li, Y.; Scolari, F.; Sanna-Cherchi, S.; Choi, M.; Verbitsky, M.; Fasel, D.A.; Lata, S.; Prakash, S.; Shapiro, S.; et al. Discovery of new risk loci for IgA nephropathy implicates genes involved in immunity against intestinal pathogens. Nat. Genet. 2014, 46, 1187–1196. [Google Scholar] [CrossRef] [PubMed]
  32. Kortman, G.A.M.; Reijnders, D.; Swinkels, D.W. Oral iron supplementation: Potential implications for the gut microbiome and metabolome in patients with CKD. Hemodial. Int. 2017, 21, S28–S36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kortman, G.A.M.; Dutilh, B.E.; Maathuis, A.J.H.; Engelke, U.F.; Boekhorst, J.; Keegan, K.P.; Nielsen, F.G.G.; Betley, J.R.; Weir, J.C.; Kingsbury, Z.; et al. Microbial Metabolism Shifts Towards an Adverse Profile with Supplementary Iron in the TIM-2 In vitro Model of the Human Colon. Front. Microbiol. 2016, 6, 1481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Satoh, M.; Hayashi, H.; Watanabe, M.; Ueda, K.; Yamato, H.; Yoshioka, T.; Motojima, M. Uremic Toxins Overload Accelerates Renal Damage in a Rat Model of Chronic Renal Failure. Nephron Exp. Nephrol. 2003, 95, e111–e118. [Google Scholar] [CrossRef]
  35. Devlin, A.S.; Marcobal, A.; Dodd, D.; Nayfach, S.; Plummer, N.; Meyer, T.; Pollard, K.S.; Sonnenburg, J.L.; Fischbach, M. Modulation of a Circulating Uremic Solute via Rational Genetic Manipulation of the Gut Microbiota. Cell Host Microbe 2016, 20, 709–715. [Google Scholar] [CrossRef] [Green Version]
  36. Meijers, B.; Bammens, B.; De Moor, B.; Verbeke, K.; Vanrenterghem, Y.; Evenepoel, P. Free p-cresol is associated with cardiovascular disease in hemodialysis patients. Kidney Int. 2008, 73, 1174–1180. [Google Scholar] [CrossRef] [Green Version]
  37. Bammens, B.; Evenepoel, P.; Keuleers, H.; Verbeke, K.; Vanrenterghem, Y. Free serum concentrations of the protein-bound retention solute p-cresol predict mortality in hemodialysis patients. Kidney Int. 2006, 69, 1081–1087. [Google Scholar] [CrossRef]
  38. Tang, W.W.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal Microbial Metabolism of Phosphatidylcholine and Cardiovascular Risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [Green Version]
  39. Tomlinson, J.A.; Wheeler, D.C. The role of trimethylamine N-oxide as a mediator of cardiovascular complications in chronic kidney disease. Kidney Int. 2017, 92, 809–815. [Google Scholar] [CrossRef] [Green Version]
  40. Shafi, T.; Powe, N.R.; Meyer, T.W.; Hwang, S.; Hai, X.; Melamed, M.L.; Banerjee, T.; Coresh, J.; Hostetter, T.H. Trimethylamine N-Oxide and Cardiovascular Events in Hemodialysis Patients. J. Am. Soc. Nephrol. 2016, 28, 321–331. [Google Scholar] [CrossRef] [Green Version]
  41. Nygård, O.; Nordrehaug, J.E.; Refsum, H.; Ueland, P.M.; Farstad, M.; Vollset, S.E. Plasma Homocysteine Levels and Mortality in Patients with Coronary Artery Disease. N. Engl. J. Med. 1997, 337, 230–237. [Google Scholar] [CrossRef] [PubMed]
  42. Ponte, B.; Pruijm, M.; Marques-Vidal, P.; Martin, P.-Y.; Burnier, M.; Paccaud, F.; Waeber, G.; Vollenweider, P.; Bochud, M. Determinants and burden of chronic kidney disease in the population-based CoLaus study: A cross-sectional analysis. Nephrol. Dial. Transplant. 2013, 28, 2329–2339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Jamison, R.L.; Hartigan, P.; Kaufman, J.S.; Goldfarb, D.S.; Warren, S.R.; Guarino, P.D.; Gaziano, J.M.; For the Veterans Affairs Site Investigators. Effect of homocysteine lowering on mortality and vascular disease in advanced chronic kidney disease and end-stage renal disease: A randomized controlled trial. JAMA 2007, 298, 1163–1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kalim, S.; Clish, C.B.; Deferio, J.J.; Ortiz, G.; Moffett, A.S.; Gerszten, R.E.; Thadhani, R.I.; Rhee, E.P. Cross-sectional examination of metabolites and metabolic phenotypes in uremia. BMC Nephrol. 2015, 16, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Pallister, T.; Jackson, M.A.; Martin, T.C.; Zierer, J.; Jennings, A.; Mohney, R.P.; MacGregor, A.; Steves, C.J.; Cassidy, A.; Spector, T.D.; et al. Hippurate as a metabolomic marker of gut microbiome diversity: Modulation by diet and relationship to metabolic syndrome. Sci. Rep. 2017, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Schulman, G.; Agarwal, R.; Acharya, M.; Berl, T.; Blumenthal, S.; Kopyt, N. A multicenter, randomized, double-blind, placebo-controlled, dose-ranging study of AST-120 (Kremezin) in patients with moderate to severe CKD. Am. J. Kidney Dis. 2006, 47, 565–577. [Google Scholar] [CrossRef]
  47. Schulman, G.; Berl, T.; Beck, G.J.; Remuzzi, G.; Ritz, E.; Arita, K.; Kato, A.; Shimizu, M. Randomized Placebo-Controlled EPPIC Trials of AST-120 in CKD. J. Am. Soc. Nephrol. 2015, 26, 1732–1746. [Google Scholar] [CrossRef] [Green Version]
  48. Gibson, G.R.; Scott, K.P.; Rastall, R.A.; Tuohy, K.M.; Hotchkiss, A.; Dubert-Ferrandon, A.; Gareau, M.; Murphy, E.F.; Saulnier, D.; Loh, G.; et al. Dietary prebiotics: Current status and new definition. Food Sci. Technol. Bull. Funct. Foods 2010, 7, 1–19. [Google Scholar] [CrossRef] [Green Version]
  49. Bliss, D.Z.; Stein, T.P.; Schleifer, C.R.; Settle, R.G. Supplementation with gum arabic fiber increases fecal nitrogen excretion and lowers serum urea nitrogen concentration in chronic renal failure patients consuming a low-protein diet. Am. J. Clin. Nutr. 1996, 63, 392–398. [Google Scholar] [CrossRef]
  50. Effect of Probiotics and Prebiotics in Renal Function in Septic Acute Kidney Injury Patients. Available online: https://ClinicalTrials.gov/show/NCT03877081 (accessed on 12 October 2020).
  51. Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A.R. Regulation of immune cell function by short-chain fatty acids. Clin. Transl. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef]
  52. Pluznick, J.L.; Protzko, R.J.; Gevorgyan, H.; Peterlin, Z.; Sipos, A.; Han, J.; Brunet, I.; Wan, L.-X.; Rey, F.; Wang, T.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Guida, B.; Germanò, R.; Trio, R.; Russo, D.; Memoli, B.; Grumetto, L.; Barbato, F.; Cataldi, M. Effect of short-term synbiotic treatment on plasma p-cresol levels in patients with chronic renal failure: A randomized clinical trial. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 1043–1049. [Google Scholar] [CrossRef] [PubMed]
  55. Rossi, M.; Johnson, D.W.; Morrison, M.; Pascoe, E.M.; Coombes, J.S.; Forbes, J.M.; Szeto, C.-C.; McWhinney, B.C.; Ungerer, J.P.; Campbell, K.L. Synbiotics Easing Renal Failure by Improving Gut Microbiology (SYNERGY): A Randomized Trial. Clin. J. Am. Soc. Nephrol. 2016, 11, 223–231. [Google Scholar] [CrossRef] [PubMed]
  56. Johnson, C.; Zhang, S.; Omede, F.; Stubbs, J. Rifaximin effects on serum trimethylamine-n-oxide in chronic kidney disease. FASEB J. 2018. [Google Scholar] [CrossRef]
  57. Kimber, C.; Zhang, S.; Johnson, C.; West, R.E., III; Prokopienko, A.J.; Mahnken, J.D.; Yu, A.S.; Hoofngale, A.N.; Ir, D.; Robertson, C.E.; et al. Randomized, Placebo-controlled Trial of Rifaximin Therapy for Lowering Gut- derived Cardiovascular Toxins and Inflammation in Chronic Kidney Disease. Kidney 2020, 360. [Google Scholar] [CrossRef]
  58. Cammarota, G.; Ianiro, G.; Tilg, H.; Rajilić-Stojanović, M.; Kump, P.; Satokari, R.; Sokol, H.; Arkkila, P.; Pintus, C.; Hart, A.; et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut 2017, 66, 569–580. [Google Scholar] [CrossRef]
  59. Gonzalez, H.U.D.J.E. Fecal Microbiota Transplantation as a Therapeutic Strategy in the Progression of Chronic Kidney Disease. 2018. Available online: https://ClinicalTrials.gov/show/NCT04361097 (accessed on 12 October 2020).
  60. Innovative Approach to Fecal Microbiota Transplantation (FMT) Applied for Chronic Kidney Disease (CKD). Available online: https://ClinicalTrials.gov/show/NCT04222153 (accessed on 12 October 2020).
  61. Mackowiak, P.A. Recycling Metchnikoff: Probiotics, the Intestinal Microbiome and the Quest for Long Life. Front. Public Health 2013, 1, 52. [Google Scholar] [CrossRef]
Figure 1. Search strategy and results.
Figure 1. Search strategy and results.
Toxins 12 00788 g001
Figure 2. Gut-kidney axis in acute kidney injury (AKI, acute kidney injury; Hcy, homocysteine; IRi, ischemia-reperfusion injury; LPS, lipopolysaccharide; TMAO, trimethylamine N-oxide).
Figure 2. Gut-kidney axis in acute kidney injury (AKI, acute kidney injury; Hcy, homocysteine; IRi, ischemia-reperfusion injury; LPS, lipopolysaccharide; TMAO, trimethylamine N-oxide).
Toxins 12 00788 g002
Figure 3. Potential microbiota-targeting interventions in acute kidney injury (AKI, acute kidney injury; FMT, fecal microbiota transplantation; OLF 78, olfactory receptor 78; ScFAs, short chain fatty acids).
Figure 3. Potential microbiota-targeting interventions in acute kidney injury (AKI, acute kidney injury; FMT, fecal microbiota transplantation; OLF 78, olfactory receptor 78; ScFAs, short chain fatty acids).
Toxins 12 00788 g003
Table 1. Original studies assessing the relationship between microbiome-derived toxins and acute kidney injury.
Table 1. Original studies assessing the relationship between microbiome-derived toxins and acute kidney injury.
AuthorYearPopulationUremic Toxin/Parameter AssessedOutcomeResults and Key Observations
Experimental
Jang et al. [5]2009germ-free mice, afterwards fed bacteria-rich dietnumbers and phenotypes of T cells and NK cells, panel of cytokinesextent of renal injury and functional decline after IRImicrobial stimuli influence the phenotype of renal lymphocytes and ameliorate the extent of renal injury
Samanta et al. [6]2017Wistar ratsmicrobiota compositionhypoxia induced AKIhypobaric hypoxia causes both AKI and affects gut microbial population
Long et al. [7]2017C57BL/6 miceinfluence of elevated Hcy levelscisplatin-induced AKIcisplatin induces more severe tubular injury, tubular cell apoptosis and lower proliferation in hyperHcy mice
Li et al. [8]2019Sprague-Dawley ratsgut-derived endotoxinincreased renal mRNA of TLR4 and proinflammatory mediators (Il-6 and MCP-1)endotoxin increases intrarenal inflammatory response
Yang et al. [9]2020C57BL/6 mice and germ-free C57BL/6 micemicrobiota compositionseverity of IRIintestinal dysbiosis, inflammation and leaky gut are consequences of AKI but also determine its severity
Andrianova et al. [10]2020Wistar ratsmicrobiota composition, levels of selected toxins (acylcarnitines)severity of IRI, creatinine and urea levelsspecific bacteria in the gut may ameliorate or aggravate IRI and affect toxin levels
Mishima et al. [11]2020germ-free mice and mice with microbiotametabolome analysisextent of kidney damage in adenine-induced AKIgerm-free mice enhanced host purine metabolism
and exacerbated kidney damage
Human
Knoflach et al. [12]1994retrospectivehippuric acid concentrationacute kidney allograft rejectionhippuric acid concentration was higher in patients with acute allograft rejection and fell after antirejection treatment
Carron et al. [13]2019prospective observational design: 146 kidney transplant recipientscirculating lipopolysaccharidechronic inflammation and acute rejection episodeschronic exposure to LPS in the period before transplantation can promote endotoxin tolerance and those patients are less prompt to develop acute rejection after transplantation
Wang et al. [14]2019prospective observational design: 262 patients with hospital-acquired AKIserum indoxyl sulfate levels90-day mortalityserum indoxyl sulfate levels were elevated in patients with AKI and associated with a worse prognosis
Veldeman et al. [15]2019prospective observational design:194 patients with sepsisserum indoxyl sulfate and p-cresyl sulfate levelsacute kidney injury due to sepsisserum indoxyl sulfate and p-cresyl sulfate levels were higher in patients with AKI and correlated with AKI course
Abbreviations: AKI, acute kidney injury; Hcy, homocysteine; Il-6, interleukin-6; IRI, ischemia-reperfusion injury; LPS, lipopolysaccharide MCP-1, monocyte chemoattractant protein-1; mRNA, messenger ribonucleic acid; TL4, toll-like receptor 4.
Table 2. Original studies assessing microbiome-modifying interventions on acute kidney injury.
Table 2. Original studies assessing microbiome-modifying interventions on acute kidney injury.
AuthorYearPopulationUremic Toxin/Parameter AssessedOutcomeResults and Key Observations
Experimental
Machado et al. [16]2012Wistar ratsSCFA (sodium butyrate)levels of creatinine, inflammatory markers and histology in contrast induced AKISCFA treatment attenuated creatinine levels and histological damage
Sun et al. [17]2013Sprague-Dawley ratsSCFA (sodium butyrate)levels of creatinine, AKI markers, antioxidant enzymes and histology in gentamicin induced AKIchronic treatment with SCFA protects from gentamicin-induced nephrotoxicity
Andrade-Oliveira et al. [18]2015C57BL/6 miceSCFAs (acetate, butyrate, propionate)levels of creatinine and urea, necrosis score in kidney tubular epithelial cells in IRImice treated with acetate-producing bacteria had improved mitochondrial biogenesis and better outcomes
Fujii et al. [19]2016SH ratsAST-120myocardial infarction induced kidney damagetreatment with AST-120 may have protective effects (reduced indole levels and urine, serum biomarkers of kidney injury)
Emal et al. [20]2017C57BL/6 wild-type micebroad-spectrum antibioticsrenal damage and tubular integrity after IRIdepletion of gut microbiota protects against renal injury
Nakade et al. [21]2018germ-free C57BL/6 miceD-serinehypoxia-induced tubular damage and kidney functionrenoprotective effects of gut-derived D-serine in AKI proven
Al-Harbi et al. [22]2018BALB/c miceSCFA (sodium acetate)kidney function/ renal peroxidase activity/kidney tubular structure in sepsis induced AKIacetate ameliorates sepsis-induced kidney injury by restoration of oxidant–antioxidant balance in T cells
Lee et al. [23]2020Sprague-Dawley rats and Caco-2 cellsLactobacillus salivarius BP121cisplatin-induced AKI occurenceL. salivarius BP121 reduced Caco-2 damage and protected against cisplatin-induced AKI
Zheng et al. [24]2020BALB/c mice and Bama miniature pigsmicrobial cocktail (Escherichia, Bacillus, Enterobacter)urea and creatinine concentration in nephrotoxin-induced AKI (adenine, cisplatin, glycerol)in both murine and porcine models of AKI the orally delivered cocktail reduced urea and creatinine concentration
Human
Dong et al. [25]2016retrospective analysis, 176 cirrhotic adult patients (88 treated with rifaximinrifaximinAKI & HRS riskincidence rate ratio of AKI and HRS, as well as the risk of RRT was lower in the rifaximin group
Abbreviations: Abbreviations: AKI, acute kidney injury; HRS, hepatorenal syndrome; IRI, ischemia-reperfusion injury; RRT, renal replacement therapy; SCFAs, short chain fatty acids; SH, spontaneously hypertensive.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rydzewska-Rosołowska, A.; Sroka, N.; Kakareko, K.; Rosołowski, M.; Zbroch, E.; Hryszko, T. The Links between Microbiome and Uremic Toxins in Acute Kidney Injury: Beyond Gut Feeling—A Systematic Review. Toxins 2020, 12, 788. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins12120788

AMA Style

Rydzewska-Rosołowska A, Sroka N, Kakareko K, Rosołowski M, Zbroch E, Hryszko T. The Links between Microbiome and Uremic Toxins in Acute Kidney Injury: Beyond Gut Feeling—A Systematic Review. Toxins. 2020; 12(12):788. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins12120788

Chicago/Turabian Style

Rydzewska-Rosołowska, Alicja, Natalia Sroka, Katarzyna Kakareko, Mariusz Rosołowski, Edyta Zbroch, and Tomasz Hryszko. 2020. "The Links between Microbiome and Uremic Toxins in Acute Kidney Injury: Beyond Gut Feeling—A Systematic Review" Toxins 12, no. 12: 788. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins12120788

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop