Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
Next Article in Journal
Lantox—The Chinese Botulinum Toxin Drug—Complete English Bibliography and Comprehensive Formalised Literature Review
Next Article in Special Issue
Gut Microbiota Dynamics and Uremic Toxins
Previous Article in Journal
Contamination by Aflatoxins B/G in Food and Commodities Imported in Southern Italy from 2017 to 2020: A Risk-Based Evaluation
Previous Article in Special Issue
Dysbiosis-Related Advanced Glycation Endproducts and Trimethylamine N-Oxide in Chronic Kidney Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Significance of the Gut Microbiota in Acute Kidney Injury

1
Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa 920-1192, Japan
2
Division of Infection Control, Kanazawa University Hospital, Kanazawa 920-1192, Japan
*
Author to whom correspondence should be addressed.
Toxins 2021, 13(6), 369; https://doi.org/10.3390/toxins13060369
Submission received: 23 March 2021 / Revised: 10 May 2021 / Accepted: 19 May 2021 / Published: 22 May 2021
(This article belongs to the Special Issue Gut Microbiota Dynamics and Uremic Toxins)

Abstract

:
Recent studies have revealed that the gut microbiota plays a crucial role in maintaining a healthy, as well as diseased condition. Various organs and systems, including the kidney, are affected by the gut microbiota. While the impacts of the gut microbiota have been reported mainly on chronic kidney disease, acute kidney injury (AKI) is also affected by the intestinal environment. In this review, we discussed the pathogenesis of AKI, highlighting the relation to the gut microbiota. Since there is no established treatment for AKI, new treatments for AKI are highly desired. Some kinds of gut bacteria and their metabolites reportedly have protective effects against AKI. Current studies provide new insights into the role of the gut microbiota in the pathogenesis of AKI.
Key Contribution: The gut microbiota is involved in the pathogenesis of acute kidney injury(AKI). The gut microbiota and its metabolites could be a novel therapeutic target for AKI.

1. Overview of the Gut Microbiota

Over 1000 genera of bacteria colonize the human intestine and shape the composition of the gut microbiota [1]. Most of the bacteria in the human intestine belong to four phyla, Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria [2]. The gut microbiota plays an important role in maintaining homeostasis in the human gastrointestinal tract. In particular, the gut microbiota contributes to mucosal immunity and nutrient metabolism in the intestine. Firstly, as a host defense mechanism, the intestinal epithelium acts as a barrier between the luminal side and the lamina propria to prevent the entry of pathogens. The intestinal epithelial cells are connected by tight junctions to form a multifunctional cell complex [3]. The gut microbiota contributes to the restoration of the protein structure of the epithelial cell tight junctions [4]. In addition, the gut microbiota is involved in the production of defense-related molecules such as heat shock proteins [5,6] and mucin genes [7] in the intestinal epithelial cells. The gut microbiota also competes with pathogenic bacteria [8] and secrete antimicrobial peptides [9] to maintain homeostasis in the intestines. Secondly, the gut microbiota regulates the host’s intestinal mucosal immune system. Segmented filamentous bacteria, a type of intestinal bacteria, are associated with the differentiation and induction of interleukin-17-producing helper T cells, which are involved in inflammatory responses [10]. Clostridiales have also been shown to contribute to the differentiation and induction of regulatory T cells [11,12]. Thirdly, the gut microbiota is also related to nutrition. Enterobacteriaceae break down indigestible carbohydrates ingested by humans and produce short-chain fatty acids (SCFA) [13], which are used as an energy source by epithelial cells [14]. The gut microbiota also synthesizes vitamin K and B vitamins [15]. Thus, the gut microbiota plays many roles. In addition, an altered composition of the gut microbiota, termed dysbiosis, is linked to disrupted homeostasis and various diseases such as inflammatory bowel disease (IBD) [16,17], irritable bowel syndrome [18,19], asthma [20], and acute kidney injury (AKI) [21,22,23,24]. Multiple factors affect the composition of the gut microbiota. Concerning external factors, living area [25], eating habits [26], exercise [27], tobacco smoking [28], beverages, and drugs [29] are associated with the composition of the gut microbiota. Genetic background, age, sex, body mass index, and race are raised as internal factors that affect the gut microbiota [30,31,32]. In a study analyzing the gut microbiota in IBD, the results differed among countries [33], suggesting that comparison under varied circumstances may lead to inconsistent results. To understand the relationship between diseases and the gut microbiota, it would be preferable to compare within groups having the same background.

2. Pathologies Associated with Acute Kidney Injury and the Gut Microbiota

The pathogenesis of AKI is composed of various factors including inflammation, apoptosis, hemodynamic changes, and oxidative stress [34]. Although there are many reports on the relationship between kidney disease and the gut microbiota, the precise mechanisms are still unknown.

2.1. The Contribution of the Gut Microbiota

Germ-free (GF) mice are often used to explore the role of the gut microbiota in the pathogenesis of AKI. Kidneys of GF mice in steady state exhibited higher number of Natural Killer T cells and lower interleukin (IL)-4 levels compared with normal mice. GF mice with ischemia-reperfusion (I/R)-induced AKI showed exacerbated renal injury and increased CD8 T cells compared with those in normal mice. The transplantation of enterobacteria from normal mice into these GF mice resulted in reduced renal dysfunction and histological damage and a change in the composition of lymphocytes in the kidneys in the I/R model [35]. These findings suggest that the presence of the gut microbiota is involved in phenotypic changes of renal lymphocytes and alleviating renal damage in I/R, and that the gut microbiota have a renoprotective aspect. We also confirmed the renoprotective effects of the gut microbiota; I/R-induced renal injury was exacerbated in GF mice compared with that in normal mice, which was alleviated by stool transplantation from normal mice into GF mice [21]. Conversely, the depletion of the gut microbiota with antimicrobial agents reportedly protected against renal damage in the murine I/R model [36]. In mice with intestinal microbiota reduced by antimicrobial agent treatment, the number of F4/80 macrophages was decreased. In addition, the expression of the chemokine receptors C-X3-C motif chemokine receptor 1 (CX3CR1) and C-C motif chemokine receptor 2 (CCR2) on kidney F4/80+ macrophages and bone marrow monocytes was decreased in these gut bacteria-depleted mice. Furthermore, stool transplantation from normal mice worsened the kidney injury in the gut microbiota-depleted mice. Since the major findings were inconsistent between the reports, further studies are needed to elucidate the impact of gut microbiota on the pathogenesis of AKI.

2.2. SCFA and Inflammation

The gut microbiota produces SCFA, such as acetate, propionate, and butyrate, from indigestible dietary fibers. They are sources of energy for the body and are also involved in inflammation. It was reported that there are four receptors (GPR41, GPR43, Oflr78, and GPR109a) for SCFA in the kidney. GPR41 and GPR43 were identified in the kidney tissue and in the renal arteries [37], and the expression of GPR43 in the kidneys was increased by acetate administration in a mouse I/R model [38,39]. GPR43 was also expressed in granulocytes, monocytes, dendritic cells, and mast cells [40,41,42], suggesting that SCFA is involved in inflammation and immune responses. Oflr78 was expressed in the vascular smooth muscle and the juxtaglomerular apparatus, which regulates renin secretion in response to SCFA [37]. In Oflr78 knockout mice, antibiotic treatment reduced the biomass of the gut microbiota and an increase in blood pressure. This suggests that SFCA from the gut microbiota modulates blood pressure via Oflr78 [37]. Although GPR109a has been reported to be present in adipose tissue in the kidney, its function in kidney disease has not been clarified [43]. GPR109a stimulates the differentiation of regulatory T cells and interleukin 10-producing T cells [44], inhibits NF-κB activity, and induces apoptosis independent of histone deacetylases inhibition [45]. These are thought to be linked to the anti-inflammatory and tumor suppressive effects of butyrate. In the I/R-induced AKI model, it was reported that acetate directly acts on dendritic cells and tubular epithelial cells via GPR43, resulting in reduced kidney injury [38]. In a mouse model of lipopolysaccharide-induced AKI, acetate administration contributed to a decrease in T-cell histone deacetylase activity and reactive oxygen species, improved renal function, and reduced tubular damage [39]. In a murine model of Adriamycin-induced renal injury, butyrate from the gut microbiota was reported to mediate gene modification via GPR109a in renal epithelial cells, thereby reducing epithelial cell damage [46]. The nephroprotective effect of butyrate (reduced tubular damage) has also been reported in a contrast-induced AKI model in rats [47]. Because most of these findings were based on animal studies, the role of SCFA in the pathogenesis of human kidney diseases remains to be investigated.

2.3. Hemodynamics

Hypertension is often accompanied by AKI. It was reported that administration of angiotensin II did not cause an increase in blood pressure in GF mice, but increased blood pressure in normal mice with gut microbiota [48]. The reduced gene expressions of vascular monocyte chemoattractant protein 1 (MCP-1), inducible nitric oxide synthase (iNOS) and NADPH oxidase subunit Nox2 were demonstrated in GF mice treated with angiotensin II compared to normal mice, as well as a reduced upregulation of retinoic-acid receptor-related orphan receptor gamma t (Rorγt), the signature transcription factor for IL-17 synthesis [48]. These results led to an attenuated vascular leukocyte adhesion, and less infiltration of Ly6G(+) neutrophils and Ly6C(+) monocytes into the aortic vessel wall [48]. This suggested that the gut microbiota is involved in vascular immune cell infiltration and inflammation in angiotensin II administered mice, leading hypertension. In addition, it was reported that high-salt-feeding (2% NaCl in drinking water) mice developed renal damage and hypertension, and that fecal transplantation from these mice into normal mice resulted in leaky gut and hypertensive renal damage [49]. These results suggested that salt loading causes dysbiosis of the intestinal microbiota, which in turn triggers renal damage. Regarding this, the SCFA receptor was reportedly expressed in the juxtaglomerular apparatus, suggesting that gut microbiota-derived SCFA is involved in blood pressure regulation. Thus, the gut microbiota is likely to be associated with blood pressure.

2.4. Uremia and Dysbiosis

In patients with end-stage renal disease (ESRD), multiple factors such as uremia, diet, and drug use alter the composition of the gut microbiota. Therefore, the components of the intestinal microbiota differ between patients with ESRD and healthy subjects [50]. Interestingly, in patients with ESRD, DNA from gut bacteria was detected in the blood in addition to increased blood levels of D-lactate, high-sensitivity C-reactive protein, and IL-6 [51]. Moreover, it was reported that plasma endotoxin/lipopolysaccharide levels were increased in accordance with the chronic kidney disease (CKD) stage [52]. These data indicated that the gut microbiota and their products enter the circulation through the intestinal barrier and affect the immune system, leading to inflammation in CKD and ESRD. Although it is clear that AKI changes the components of the gut microbiota and its metabolites (Table 1), the precise mechanisms for the alteration of the gut microbiota in AKI need to be clarified.

2.5. Septic AKI

Sepsis is one of the most common risks for the development of AKI. 45 to 70% of AKI is reportedly associated with sepsis [53]. Septic AKI also alters composition of the gut microbiota, especially the loss of anaerobic bacteria. Systemic inflammation, antibiotics treatment, parenteral nutrition, and leaky gut may cause dysbiosis of the gut microbiota. In addition, decreased the gut microbiota and parenteral nutrition reduce the production of SCFA, which function as immune modulators [54].

3. Products Derived from the Gut Microbiota and AKI

3.1. D-Amino Acids

Amino acids have optical isomers, which can be distinguished into L- and D-forms, called chiral amino acids. In recent years, the technical innovation for measuring amino acids enabled us to separate and measure the chiral L- and D-forms with high accuracy [55]. The distributions and functions of D-amino acids were unclear until recently. In turn, chiral amino acid analysis revealed the existence of D-forms in vivo, and the kinetics and functions of D-amino acids in the body are now highlighted. In animal models, D-amino acid synthases and degrading enzymes are abundant in the brain and kidneys [56,57], suggesting that D-amino acids are metabolized in these organs. We have found that the stool of normal mice contains higher amounts of D-amino acids than that in the stool of GF mice [21], suggesting that D-amino acids are produced by the gut microbiota. In addition, we found that D-serine from intestinal bacteria showed reno-protective effects in a murine AKI model [21]. Nevertheless, a high concentration of D-serine induces renal damage [21,58,59], suggesting that the amount of D-serine is associated with specific responses, especially in the kidney. In addition to D-serine, various other kinds of D-amino acids are distributed in the organs. The pathophysiological roles of D-amino acids need to be elucidated.

3.2. Uremic Substances

Indoxyl sulfate (IS), p-cresyl sulfate (p-CS), and trimethylamine-N-oxide (TMAO) are known to be uremic substances associated with the gut microbiota. Studies with GF mice revealed that IS, p-CS, and TMAO are derived from the gut microbiota, and that diet is another source of TMAO [60]. The toxicity of IS and p-CS has been well studied in CKD, and their blood levels were associated with cardiovascular events and mortality in dialysis patients [61,62]. Similarly, elevated blood levels of TMAO were associated with the risk of cardiovascular disease and death [63]. In obese mouse models, TMAO-induced renal interstitial fibrosis was also reported [64]. It was reported that CKD patients with high blood TMAO levels had a poor life prognosis [65], and that dialysis patients with high TMAO serum levels had a higher risk of coronary events and death [66]. Additionally, recent systematic review and meta-analysis revealed a negative association between circulating TMAO concentrations and kidney function in CKD patients [67]. Uremic substances accumulated even in the AKI condition. The serum IS level was higher in AKI patients who required hospitalization than that in healthy subjects [68]. In this study, the mortality was increased in the group with higher serum IS levels [68]. Another report showed that serum IS and p-CS levels were elevated in patients with AKI and sepsis [69]. In both reports, it was also observed that serum levels of IS and p-CS were lower in AKI than in CKD [68,69]. Further studies are needed to elucidate the pathophysiological role of uremic substances in AKI.

4. Gut Microbiota as a Potential Therapeutic Option for AKI

Although some reports have raised gut microbiota and its products as therapeutic targets for CKD, the therapeutic potential still remains to be investigated in AKI patients. A recent systematic review shows that modifying the gut microbiota improves the prognosis of AKI [70]. Among the metabolites of the gut microbiota, SCFA and D-serine reportedly have renoprotective effects in AKI. These metabolites could be therapeutic options as postbiotics. In turn, some reports showed the therapeutic effects of probiotics. Lactobacillus salivarius BP121 attenuated cisplatin-induced tubular injury concomitant with reduced inflammation and oxidative stress [71]. A microbial cocktail of Escherichia, Bacillus, and Enterobacter reportedly protected against nephrotoxin-induced AKI [72]. ‘Prebiotics’ is a general term for nondigestible food ingredients (such as dietary fiber) that promote the growth of gut microbiota. Concerning prebiotics, their therapeutic effect in AKI has not been reported yet. However, a randomized clinical trial of probiotics and prebiotics in septic AKI was conducted [73], but the result is still not published. It was also reported that a high-fiber diet suppressed acute allograft rejection in a kidney transplant model [74].
Adsorbents are used to reduce uremic substances derived from the gut microbiota. AST-20 is effective in adsorbing precursors of uremic toxins such as indole and p-cresol. Although AST-20 reportedly reduced the decline of kidney function in CKD, the renoprotective effect in AKI is not clear. However, the lowering of uremic substances seems to have benefits even in AKI, based on the observational studies. Elevated levels of IS and p-CS in AKI were correlated with the severity of AKI (RIFLE classification) [75], and IS levels were associated with mortality in AKI, requiring hospitalization [68]. The suppression of IS and p-CS in the study of Lactobacillus salivarius BP121 mentioned previously suggested that the suppression or adsorption of these uremic substances in AKI may be a therapeutic target. In addition, rifaximin, a kind of nonabsorbable antibiotic, decreased the TMAO blood level in a mouse model of CKD [76]. Moreover, rifaximin treatment reduced the incidence of AKI, hepatorenal syndrome, and the need for renal replacement therapy in a human retrospective study [77]. The pathophysiologic roles of uremic substances need to be investigated in the AKI condition.

5. Conclusions

The relationship between AKI and the gut microbiota is still poorly understood (Figure 1), unlike the relationship between CKD and the gut microbiota. Further studies on AKI pathogenesis and therapeutic applications are expected.

Author Contributions

Writing—original draft preparation, T.K., Y.I., and Y.N.; writing—review and editing, T.K., Y.I., and T.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This review was supported by AMED under Grant Number JP20ek0310012.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gilbert, J.A.; Blaser, M.J.; Caporaso, J.G.; Jansson, J.K.; Lynch, S.V.; Knight, R. Current understanding of the human microbiome. Nat. Med. 2018, 24, 392–400. [Google Scholar] [CrossRef]
  2. Frank, D.N.; St Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Farquhar, M.G.; Palade, G.E. Junctional complexes in various epithelia. J. Cell. Biol. 1963, 17, 375–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ewaschuk, J.B.; Diaz, H.; Meddings, L.; Diederichs, B.; Dmytrash, A.; Backer, J.; Looijer-van Langen, M.; Madsen, K.L. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G1025–G1034. [Google Scholar] [CrossRef] [Green Version]
  5. Fujiya, M.; Musch, M.W.; Nakagawa, Y.; Hu, S.; Alverdy, J.; Kohgo, Y.; Schneewind, O.; Jabri, B.; Chang, E.B. The Bacillus subtilis quorum-sensing molecule CSF contributes to intestinal homeostasis via OCTN2, a host cell membrane transporter. Cell Host Microbe 2007, 1, 299–308. [Google Scholar] [CrossRef] [Green Version]
  6. Tao, Y.; Drabik, K.A.; Waypa, T.S.; Musch, M.W.; Alverdy, J.C.; Schneewind, O.; Chang, E.B.; Petrof, E.O. Soluble factors from Lactobacillus GG activate MAPKs and induce cytoprotective heat shock proteins in intestinal epithelial cells. Am. J. Physiol. Cell Physiol. 2006, 290, C1018–C1030. [Google Scholar] [CrossRef] [Green Version]
  7. Mattar, A.F.; Teitelbaum, D.H.; Drongowski, R.A.; Yongyi, F.; Harmon, C.M.; Coran, A.G. Probiotics up-regulate MUC-2 mucin gene expression in a Caco-2 cell-culture model. Pediatr. Surg. Int. 2002, 18, 586–590. [Google Scholar]
  8. Sherman, P.M.; Johnson-Henry, K.C.; Yeung, H.P.; Ngo, P.S.; Goulet, J.; Tompkins, T.A. Probiotics reduce enterohemorrhagic Escherichia coli O157:H7-and enteropathogenic E. coli O127:H6-induced changes in polarized T84 epithelial cell monolayers by reducing bacterial adhesion and cytoskeletal rearrangements. Infect. Immun. 2005, 73, 5183–5188. [Google Scholar] [CrossRef] [Green Version]
  9. Schlee, M.; Harder, J.; Koten, B.; Stange, E.F.; Wehkamp, J.; Fellermann, K. Probiotic lactobacilli and VSL#3 induce enterocyte beta-defensin 2. Clin. Exp. Immunol. 2008, 151, 528–535. [Google Scholar]
  10. Ivanov, I.I.; Atarashi, K.; Manel, N.; Brodie, E.L.; Shima, T.; Karaoz, U.; Wei, D.; Goldfarb, K.C.; Santee, C.A.; Lynch, S.V.; et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009, 139, 485–498. [Google Scholar] [CrossRef] [Green Version]
  11. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–341. [Google Scholar] [CrossRef] [Green Version]
  12. Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef]
  13. Topping, D.L.; Clifton, P.M. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 2001, 81, 1031–1064. [Google Scholar] [CrossRef]
  14. Pomare, E.W.; Branch, W.J.; Cummings, J.H. Carbohydrate fermentation in the human colon and its relation to acetate concentrations in venous blood. J. Clin. Investig. 1985, 75, 1448–1454. [Google Scholar] [CrossRef] [Green Version]
  15. Hill, M.J. Intestinal flora and endogenous vitamin synthesis. Eur. J. Cancer Prev. 1997, 6 (Suppl. 1), S43–S45. [Google Scholar] [CrossRef]
  16. Andoh, A.; Sakata, S.; Koizumi, Y.; Mitsuyama, K.; Fujiyama, Y.; Benno, Y. Terminal restriction fragment length polymorphism analysis of the diversity of fecal microbiota in patients with ulcerative colitis. Inflam. Bowel Dis. 2007, 13, 955–962. [Google Scholar] [CrossRef]
  17. Andoh, A.; Kuzuoka, H.; Tsujikawa, T.; Nakamura, S.; Hirai, F.; Suzuki, Y.; Matsui, T.; Fujiyama, Y.; Matsumoto, T. Multicenter analysis of fecal microbiota profiles in Japanese patients with Crohn’s disease. J. Gastroenterol. 2012, 47, 1298–1307. [Google Scholar] [CrossRef]
  18. Krogius-Kurikka, L.; Lyra, A.; Malinen, E.; Aarnikunnas, J.; Tuimala, J.; Paulin, L.; Makivuokko, H.; Kajander, K.; Palva, A. Microbial community analysis reveals high level phylogenetic alterations in the overall gastrointestinal microbiota of diarrhoea-predominant irritable bowel syndrome sufferers. BMC Gastroenterol. 2009, 9, 95. [Google Scholar] [CrossRef] [Green Version]
  19. Carroll, I.M.; Chang, Y.H.; Park, J.; Sartor, R.B.; Ringel, Y. Luminal and mucosal-associated intestinal microbiota in patients with diarrhea-predominant irritable bowel syndrome. Gut Pathog. 2010, 2, 19. [Google Scholar] [CrossRef] [Green Version]
  20. Durack, J.; Kimes, N.E.; Lin, D.L.; Rauch, M.; McKean, M.; McCauley, K.; Panzer, A.R.; Mar, J.S.; Cabana, M.D.; Lynch, S.V. Delayed gut microbiota development in high-risk for asthma infants is temporarily modifiable by Lactobacillus supplementation. Nat. Commun. 2018, 9, 707. [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, e97957. [Google Scholar] [CrossRef] [Green Version]
  22. Yang, J.; Kim, C.J.; Go, Y.S.; Lee, H.Y.; Kim, M.G.; Oh, S.W.; Cho, W.Y.; Im, S.H.; Jo, S.K. Intestinal microbiota control acute kidney injury severity by immune modulation. Kidney Int. 2020, 98, 932–946. [Google Scholar] [CrossRef]
  23. Andrianova, N.V.; Popkov, V.A.; Klimenko, N.S.; Tyakht, A.V.; Baydakova, G.V.; 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] [PubMed] [Green Version]
  24. Samanta, A.; Patra, A.; Mandal, S.; Roy, S.; Das, K.; Kar, S.; Nandi, D.K. Hypoxia: A cause of acute renal failure and alteration of gastrointestinal microbial ecology. Saudi J. Kidney Dis. Transpl. 2018, 29, 879–888. [Google Scholar]
  25. He, Y.; Wu, W.; Zheng, H.M.; Li, P.; McDonald, D.; Sheng, H.F.; Chen, M.X.; Chen, Z.H.; Ji, G.Y.; Zheng, Z.D.; et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 2018, 24, 1532–1535. [Google Scholar] [CrossRef]
  26. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [Green Version]
  28. Savin, Z.; Kivity, S.; Yonath, H.; Yehuda, S. Smoking and the intestinal microbiome. Arch. Microbiol. 2018, 200, 677–684. [Google Scholar] [CrossRef]
  29. Falony, G.; Joossens, M.; Vieira-Silva, S.; Wang, J.; Darzi, Y.; Faust, K.; Kurilshikov, A.; Bonder, M.J.; Valles-Colomer, M.; Vandeputte, D.; et al. Population-level analysis of gut microbiome variation. Science 2016, 352, 560–564. [Google Scholar] [CrossRef]
  30. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; FitzGerald, M.G.; Fulton, R.S.; et al. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar]
  31. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [Green Version]
  32. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef]
  33. Rehman, A.; Rausch, P.; Wang, J.; Skieceviciene, J.; Kiudelis, G.; Bhagalia, K.; Amarapurkar, D.; Kupcinskas, L.; Schreiber, S.; Rosenstiel, P.; et al. Geographical patterns of the standing and active human gut microbiome in health and IBD. Gut 2016, 65, 238–248. [Google Scholar] [CrossRef] [PubMed]
  34. Bellomo, R.; Kellum, J.A.; Ronco, C. Acute kidney injury. Lancet 2012, 380, 756–766. [Google Scholar] [CrossRef]
  35. 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. Renal Physiol. 2009, 297, F1457–F1465. [Google Scholar] [CrossRef] [Green Version]
  36. 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. 2017, 28, 1450–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. 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] [Green Version]
  38. 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]
  39. 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]
  40. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
  41. Cox, M.A.; Jackson, J.; Stanton, M.; Rojas-Triana, A.; Bober, L.; Laverty, M.; Yang, X.; Zhu, F.; Liu, J.; Wang, S.; et al. Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines. World J. Gastroenterol. 2009, 15, 5549–5557. [Google Scholar] [CrossRef] [PubMed]
  42. Karaki, S.; Tazoe, H.; Hayashi, H.; Kashiwabara, H.; Tooyama, K.; Suzuki, Y.; Kuwahara, A. Expression of the short-chain fatty acid receptor, GPR43, in the human colon. J. Mol. Histol. 2008, 39, 135–142. [Google Scholar] [CrossRef]
  43. Mielenz, M.; Kuhla, B.; Hammon, H.M. Abundance of adiponectin system and G-protein coupled receptor GPR109A mRNA in adipose tissue and liver of F2 offspring cows of Charolais x German Holstein crosses that differ in body fat accumulation. J. Dairy Sci. 2013, 96, 278–289. [Google Scholar] [CrossRef] [PubMed]
  44. Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Thangaraju, M.; Cresci, G.A.; Liu, K.; Ananth, S.; Gnanaprakasam, J.P.; Browning, D.D.; Mellinger, J.D.; Smith, S.B.; Digby, G.J.; Lambert, N.A.; et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. 2009, 69, 2826–2832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Felizardo, R.J.F.; de Almeida, D.C.; Pereira, R.L.; Watanabe, I.K.M.; Doimo, N.T.S.; Ribeiro, W.R.; Cenedeze, M.A.; Hiyane, M.I.; Amano, M.T.; Braga, T.T.; et al. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms. FASEB J. 2019, 33, 11894–11908. [Google Scholar] [CrossRef] [Green Version]
  47. Machado, R.A.; Constantino Lde, 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]
  48. Karbach, S.H.; Schonfelder, T.; Brandao, I.; Wilms, E.; Hormann, N.; Jackel, S.; Schuler, R.; Finger, S.; Knorr, M.; Lagrange, J.; et al. Gut microbiota promote angiotensin II-induced arterial hypertension and vascular dysfunction. J. Am. Heart Assoc. 2016, 5, e003698. [Google Scholar] [CrossRef] [Green Version]
  49. Hu, J.; Luo, H.; Wang, J.; Tang, W.; Lu, J.; Wu, S.; Xiong, Z.; Yang, G.; Chen, Z.; Lan, T.; et al. Enteric dysbiosis-linked gut barrier disruption triggers early renal injury induced by chronic high salt feeding in mice. Exp. Mol. Med. 2017, 49, e370. [Google Scholar] [CrossRef] [Green Version]
  50. Vaziri, N.D.; Wong, J.; Pahl, M.; 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] [Green Version]
  51. Wang, F.; Jiang, H.; Shi, K.; Ren, Y.; Zhang, P.; Cheng, S. Gut bacterial translocation is associated with microinflammation in end-stage renal disease patients. Nephrology 2012, 17, 733–738. [Google Scholar] [CrossRef] [PubMed]
  52. McIntyre, C.W.; Harrison, L.E.; Eldehni, M.T.; Jefferies, H.J.; Szeto, C.C.; John, S.G.; Sigrist, M.K.; Burton, J.O.; Hothi, D.; Korsheed, S.; et al. Circulating endotoxemia: A novel factor in systemic inflammation and cardiovascular disease in chronic kidney disease. Clin. J. Am. Soc. Nephrol. 2011, 6, 133–141. [Google Scholar] [CrossRef] [PubMed]
  53. Bagshaw, S.M.; Laupland, K.B.; Doig, C.J.; Mortis, G.; Fick, G.H.; Mucenski, M.; Godinez-Luna, T.; Svenson, L.W.; Rosenal, T. Prognosis for long-term survival and renal recovery in critically ill patients with severe acute renal failure: A population-based study. Crit. Care 2005, 9, R700–R709. [Google Scholar] [CrossRef] [Green Version]
  54. Zhang, J.; Ankawi, G.; Sun, J.; Digvijay, K.; Yin, Y.; Rosner, M.H.; Ronco, C. Gut-kidney crosstalk in septic acute kidney injury. Crit. Care 2018, 22, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Koga, R.; Miyoshi, Y.; Negishi, E.; Kaneko, T.; Mita, M.; Lindner, W.; Hamase, K. Enantioselective two-dimensional high-performance liquid chromatographic determination of N-methyl-D-aspartic acid and its analogues in mammals and bivalves. J. Chromatogr. A 2012, 1269, 255–261. [Google Scholar] [CrossRef] [PubMed]
  56. Miyoshi, Y.; Oyama, T.; Itoh, Y.; Hamase, K. Enantioselective two-dimensional high-performance liquid chromatographic determination of amino acids: Analysis and physiological significance of d-amino acids in mammals. Chromatography 2014, 35, 49–57. [Google Scholar] [CrossRef] [Green Version]
  57. Horio, M.; Kohno, M.; Fujita, Y.; Ishima, T.; Inoue, R.; Mori, H.; Hashimoto, K. Levels of D-serine in the brain and peripheral organs of serine racemase (Srr) knock-out mice. Neurochem. Int. 2011, 59, 853–859. [Google Scholar] [CrossRef]
  58. Ganote, C.E.; Peterson, D.R.; Carone, F.A. The nature of D-serine-induced nephrotoxicity. Am. J. Pathol. 1974, 77, 269–282. [Google Scholar]
  59. Okada, A.; Nangaku, M.; Jao, T.M.; Maekawa, H.; Ishimono, Y.; Kawakami, T.; Inagi, R. D-serine, a novel uremic toxin, induces senescence in human renal tubular cells via GCN2 activation. Sci. Rep. 2017, 7, 11168. [Google Scholar] [CrossRef]
  60. Mishima, E.; Fukuda, S.; Mukawa, C.; Yuri, A.; Kanemitsu, Y.; Matsumoto, Y.; Akiyama, Y.; Fukuda, N.N.; Tsukamoto, H.; Asaji, K.; et al. Evaluation of the impact of gut microbiota on uremic solute accumulation by a CE-TOFMS-based metabolomics approach. Kidney Int. 2017, 92, 634–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Meijers, B.K.; 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] [PubMed] [Green Version]
  62. 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] [PubMed]
  63. Obeid, R.; Awwad, H.M.; Rabagny, Y.; Graeber, S.; Herrmann, W.; Geisel, J. Plasma trimethylamine N-oxide concentration is associated with choline, phospholipids, and methyl metabolism. Am. J. Clin. Nutr. 2016, 103, 703–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sun, G.; Yin, Z.; Liu, N.; Bian, X.; Yu, R.; Su, X.; Zhang, B.; Wang, Y. Gut microbial metabolite TMAO contributes to renal dysfunction in a mouse model of diet-induced obesity. Biochem. Biophys. Res. Commun. 2017, 493, 964–970. [Google Scholar] [CrossRef]
  65. Tomlinson, J.A.P.; 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]
  66. 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. 2017, 28, 321–331. [Google Scholar] [CrossRef] [Green Version]
  67. Zeng, Y.; Guo, M.; Fang, X.; Teng, F.; Tan, X.; Li, X.; Wang, M.; Long, Y.; Xu, Y. Gut Microbiota-Derived Trimethylamine N-Oxide and Kidney Function: A Systematic Review and Meta-Analysis. Adv. Nutr. 2021, nmab010. [Google Scholar] [CrossRef]
  68. 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, 57. [Google Scholar] [CrossRef]
  69. 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]
  70. Rydzewska-Rosolowska, A.; Sroka, N.; Kakareko, K.; Rosolowski, 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. [Google Scholar] [CrossRef]
  71. Lee, T.H.; Park, D.; Kim, Y.J.; Lee, I.; Kim, S.; Oh, C.T.; Kim, J.Y.; 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]
  72. 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]
  73. Effect of Probiotics and Prebiotics in Renal Function in Septic Acute Kidney Injury Patients. Available online: https://clinicaltrials.gov/ct2/show/NCT03877081 (accessed on 31 December 2020).
  74. Wu, H.; Singer, J.; Kwan, T.K.; Loh, Y.W.; Wang, C.; Tan, J.; Li, Y.J.; Lai, S.W.C.; Macia, L.; Alexander, S.I.; et al. Gut Microbial Metabolites Induce Donor-Specific Tolerance of Kidney Allografts through Induction of T Regulatory Cells by Short-Chain Fatty Acids. J. Am. Soc. Nephrol. 2020, 31, 1445–1461. [Google Scholar] [CrossRef] [PubMed]
  75. Devlin, A.S.; Marcobal, A.; Dodd, D.; Nayfach, S.; Plummer, N.; Meyer, T.; Pollard, K.S.; Sonnenburg, J.L.; Fischbach, M.A. Modulation of a circulating uremic solute via rational genetic manipulation of the gut microbiota. Cell Host Microbe 2016, 20, 709–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Johnson, C.; Zhang, S.; Omede, F.; Stubbs, J. Rifaximin effects on serum trimethylamine-n-oxide in chronic kidney disease—Johnson. FASEB J. 2018. [Google Scholar]
  77. Dong, T.; Aronsohn, A.; Gautham Reddy, K.; 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]
Figure 1. A brief summary of AKI and the gut microbiota. The gut microbiota is involved in the pathogenesis of AKI. SCFA and D-serine, products of the gut microbiota, have renoprotective effects against AKI. IS and p-CS are elevated in AKI, but their effects on AKI are unknown.
Figure 1. A brief summary of AKI and the gut microbiota. The gut microbiota is involved in the pathogenesis of AKI. SCFA and D-serine, products of the gut microbiota, have renoprotective effects against AKI. IS and p-CS are elevated in AKI, but their effects on AKI are unknown.
Toxins 13 00369 g001
Table 1. AKI model and gut microbiota analysis.
Table 1. AKI model and gut microbiota analysis.
AuthorType of AnimalAKI MethodBacterial Collection Method, PeriodBacterial Storage MethodBacterial Analysis MethodsChanged BacteriaChanged Metabolites
Nakade [21]C57BL/6 miceI/R
Unilateral 40 min
Feces
Day 0, 2, 10 after I/R
−80 °C freezer16S rRNA gene-sequencing
analysis
Increased
Lactobacillus,
Clostridium,
Ruminococcus
Decreased
Bifidobacterium
TM7
Increased
D-Serine/L-Serine
Yang [22]C57BL/6 miceI/R
Bilateral
25.5 min
(SPF mice)
28.5 min
(GF mice)
Feces
Day 1, 3, 7 after I/R
−70 °C freezer16S rRNA gene-sequencing
analysis
Increased
Enterobacteriacea
Decreased
Lactobacilli
Ruminococacceae
Decreased
SCFA
Andrianova [23]Wistar ratsI/R
Unilateral
40 min
Faces
Period is not mentioned
Not mentionedMetagenomic analysisIncreased
Staoh vlococc
Prevotella
Increased
32 acylcarnitines
Decreased
tyrosine, tryptophan, proline
Samanta [24]Wistar ratsHypoxia environment Barometric pressure: 7.3, 9.3, 11.8 psiaFeces
Day0, 7 after hypoxia
CulturedLimited bacterial analysisIncreased
Escherichia coli
Bacteroidetes
Bifidobacterium
Salmonella
Not mentioned
Abbreviations: AKI, acute kidney injury; GF, germ free; I/R, ischemia reperfusion; SPF, specific pathogen free.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kobayashi, T.; Iwata, Y.; Nakade, Y.; Wada, T. Significance of the Gut Microbiota in Acute Kidney Injury. Toxins 2021, 13, 369. https://doi.org/10.3390/toxins13060369

AMA Style

Kobayashi T, Iwata Y, Nakade Y, Wada T. Significance of the Gut Microbiota in Acute Kidney Injury. Toxins. 2021; 13(6):369. https://doi.org/10.3390/toxins13060369

Chicago/Turabian Style

Kobayashi, Taku, Yasunori Iwata, Yusuke Nakade, and Takashi Wada. 2021. "Significance of the Gut Microbiota in Acute Kidney Injury" Toxins 13, no. 6: 369. https://doi.org/10.3390/toxins13060369

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