Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Pediatr Nephrol

DOI 10.1007/s00467-016-3527-x

REVIEW

The gut–kidney axis


Pieter Evenepoel 1,2,3 & Ruben Poesen 1,2 & Björn Meijers 1,2

Received: 3 May 2016 / Revised: 2 September 2016 / Accepted: 2 September 2016


# IPNA 2016

Abstract The host–gut microbiota interaction has been the These co-metabolites are an appealing target for adjuvant ther-
focus of increasing interest in recent years. It has been deter- apy in CKD. Treatment options include dietary therapy, pre-
mined that this complex interaction is not only essential to biotics, probiotics and host and bacterial enzyme inhibitors.
many aspects of normal Bmammalian^ physiology but that it Final proof of the concept should come from randomized con-
may also contribute to a multitude of ailments, from the obvi- trolled and adequately powered intervention studies.
ous case of inflammatory bowel disease to (complex) diseases
residing in organs outside the gut. An increasing body of ev- Keywords Microbiome . Microbial metabolism .
idence indicates that crosstalk between host and microbiota is Host–gut microbiota interaction . Chronic kidney disease .
pathophysiologically relevant in patients with chronic kidney Uremic toxins
disease (CKD). Interactions are bidirectional; on the one hand,
uremia affects both the composition and metabolism of the gut
microbiota and, on the other hand, important uremic toxins Introduction
originate from microbial metabolism. In addition, gut
dysbiosis may induce a disruption of the epithelial barrier, Gut microbiota and co-metabolism
ultimately resulting in increased exposure of the host to endo-
toxins. Due to dietary restrictions and gastrointestinal dys- The human intestinal tract, and especially the large intestine, is
functions, microbial metabolism shifts to a predominantly colonized by trillions of microbes, which collectively possess
proteolytic fermentation pattern in CKD. Indoxyl sulfate and many hundredfold more genes than included in the human
p-cresyl sulfate, both end-products of protein fermentation, genome. Humans may thus be viewed as Bsupra-organisms^
and trimethylamine-N-oxide, an end-product of microbial or Bmeta-organisms^. The combined genetic potential of the
choline and carnitine metabolism, are prototypes of uremic endogenous flora is referred to as the ‘microbiome’ [1].
toxins originating from microbial metabolism. The vascular Microbiota contribute substantially to the metabolic pheno-
and renal toxicity of these co-metabolites has been demon- type of humans. Microbial metabolism, commonly referred
strated extensively in experimental and clinical studies. to as co-metabolism, comprises the synthesis of vitamins
and the fermentation of carbohydrates, lipids, proteins and bile
acids. The composition and activity of the gut microbiota co-
* Pieter Evenepoel develops with the host from birth and is subject to a complex
Pieter.Evenepoel@uzleuven.be interplay between host genome, nutrition and life-style factors
[2]. The microbiota of the infant is seeded at birth and is
1
Laboratory of Nephrology, Department of Immunology and initially undifferentiated across the various body habitats. A
Microbiology, KU Leuven, Leuven, Belgium variety of factors, including method of delivery (i.e. vaginal
2
Department of Nephrology and Renal Transplantation, University vs. Cesarean section), breast feeding and weaning, influence
Hospitals Leuven, Leuven, Belgium the infant microbiota. During suckling, the microbial commu-
3
Dienst Nefrologie, University Hospitals Leuven–Gasthuisberg nity develops rapidly; shifts in microbial diversity occur
campus, Herestraat 49, 3000 Leuven, Belgium throughout childhood and adult life; and in old age, there is
Pediatr Nephrol

a decrease in Bacteroidetes species and an increase in diffusing in intestinal contents. The colonic fate of α-amino
Firmicutes species. Extrinsic environmental and life-style fac- nitrogen (amino acids and intermediates) largely depends on
tors (such as antibiotic use, diet, stress, disease, and injury) the amount of energy available for bacterial growth and cell
continually influence the diversity and function of the gut division. The main source of energy is fermentable CHO. In
microbiota with implications for human health [1]. the case of CHO excess, α-amino nitrogen is predominantly
Among the life-style factors, diet is increasingly being ac- incorporated in the bacterial biomass; conversely, in case of
knowledged as a key regulator of intestinal microbiota and its CHO deprivation, α-amino nitrogen will be predominantly
activity [3–7] (Fig. 1). Amounts of nutrients entering the colon fermented [4] to phenols, indoles, amines, among other end-
mainly depend on dietary intake(s) and on the efficiency of the products. It is believed that fat does not reach the colonic
assimilation process in the small intestine [8]. Dietary carbo- microbiota in significant amounts as it is mostly digested
hydrates (CHO) resistant to digestion in the small intestine, and absorbed in the small intestine. Dietary fat can have an
i.e. dietary fibers, are the main source of CHO to the colon. indirect effect on the colonic microenvironment as bile acids,
CHOs are fermented to short chain fatty acids (SCFAs), which which are excreted in the small intestine to aid the digestion of
play an important role in maintaining energy homeostasis and fat, can reach the colon and be converted by the microbiota.
gut epithelial integrity [9]. Nitrogen is provided to the large Foods rich in fats are moreover often rich in the dietary nutri-
intestine by dietary proteins which escape digestion in the ents phosphatidylcholine (lecithin), choline and carnitine,
upper gut, by endogenous proteins (pancreatic and intestinal which can be used by gut microbiota as a carbon fuel source.
secretions, sloughed epithelial cells) and by blood urea While mammals do not have the enzyme that can cleave the

Fig. 1 The diet–gut–kidney axis. Following dietary exposures of certain flavin monooxygenase enzyme family to trimethylamine-N-oxide
nutrients, gut microbiota can elicit both metabolism-dependent and (TMAO) in the liver. SCFAs confer health benefits, while protein fermen-
metabolism-independent effects on the host. Metabolism-dependent ef- tation metabolites and TMAO exert multiple toxic effects, contributing to
fects include: (1) microbial fermentation of dietary carbohydrates (CHO) cardiovascular disease (CVD) and progressive renal disease (CKD), ulti-
to generate short chain fatty acids (SCFAs) which signal the host to in- mately culminating in premature death. Metabolism-independent effects
crease energy expenditure, inhibit histone deacetylase (HDAC) activity include gut hyperpermeability (leaky gut), allowing bacterial cell-wall
and enhance G protein–coupled receptor (GPCR) signaling; (2) microbial products such as lipopolysaccharide (LPS) and peptidoglycans to enter
conversion of alpha-NH3 nitrogen to protein fermentation metabolites, the bloodstream. Low circulating levels of these bacterial components
including phenols and indoles, which are subsequently metabolized in collectively activate macrophages, which can reduce reverse cholesterol
the colonocytes and liver to p-cresyl sulfate (PCS) and indoxyl sulfate transport and increase insulin resistance, hyperlipidemia and vascular
(indS), among others; (3) microbial conversion of choline and L-carnitine inflammation. LPS and peptidoglycans contribute to a micro-
to trimethylamine (TMA), which is subsequently converted by the host inflammatory state
Pediatr Nephrol

C–N bond of these nutrients, gut microbes possess hemodialysis (HD) and found no significant difference in
trimethylamine (TMA) lyases, which are able to cleave the the total number of bacteria between the patient group and
C–N bond, releasing the TMA moiety as a waste product [10]. the healthy controls [14]. However, in terms of individual
Another important and potentially modifiable determinant species, they did observe several quantitative and qualita-
of colonic microbiota composition and activity is transit time. tive changes. More specifically, the number of aerobic bac-
In a landmark study, Cummings et al. observed a significant teria, such as enterobacteria and enterococci, was about
correlation between colonic transit time and the urinary excre- 100-fold higher in the HD patients than in the healthy con-
tion rate of phenols [11]. In this study, 64 % of the variance of trols, and the number of the anaerobes Bifidobacteria spe-
the urinary excretion rate of phenols was explained by the cies and Clostridium perfringens was significantly lower
colonic transit time and dietary protein intake, with roughly and higher, respectively, in the HD patients. In another
a doubling of the colonic transit time corresponding to a 60 % study, Wang et al. analyzed the microbial composition of
increase of the urinary phenol excretion intake [11]. Also, in a patients on peritoneal dialysis by real-time PCR and found
cohort of 53 children with autism spectrum disorder, increased lower levels of Bifidobacteria species and Klebsiella
urinary excretion of p-cresol was observed in children with pneumonia [15]. Vaziri et al. recently isolated microbial
increased intestinal transit time and chronic constipation [12]. DNA from the stools of 24 patients on maintenance HD
A slowing down in colonic transit time induces an upstream and 12 healthy persons and analyzed these samples by
expansion in the number of proteolytic species, as a larger part phylogenetic microarray [16]. They observed marked dif-
of the colon becomes carbohydrate deprived, resulting in an ferences in the abundance of 190 bacterial operational tax-
increased generation and uptake of end-products of bacterial onomic units (OTUs) between the HD patients and healthy
protein fermentation. controls. To isolate the effect of uremia from inter-
individual variations, comorbid conditions and dietary
Influence of gut microbiota on health and disease and medicinal interventions (especially antibiotics),
Yoshifuji et al. studied rats at 8 weeks post 5/6 nephrecto-
The host–microbiota interaction has been the focus of increas- my or post sham operation [17]. These authors found a
ing interest in recent years. It has become clear that host–mi- significant difference in the abundance of 175 bacterial
crobe interactions are essential to many aspects of normal OTUs between the uremic and control animals, of which
Bmammalian^ physiology, ranging from nutrition to immune decreases in the Lactobacillaceae were the most notable.
homeostasis. Disturbance of the gut flora (referred to as ‘gut Thus, it would appear that uremia profoundly alters the
dysbiosis’) may contribute to a multitude of ailments, ranging composition of the gut microbiome. Additional studies by
from the obvious case of inflammatory bowel disease to com- Vaziri et al. demonstrated that patients with CKD stage 5D
plex diseases residing in organs outside the gut [13]. The notion had increased numbers of bacteria that possess urease,
that microbiota may portend disease is not new—as early as uricase, p-cresol and indole-forming enzymes and reduced
400 BC Hippocrates was quoted as saying, Bdeath sits in the numbers of bacteria that possess SCFA-forming enzymes
bowel^ and Bbad digestion is the root of all evil^. Also in the [18]. It is possible that in addition to changes in nutrient
setting of chronic kidney disease (CKD) there is mounting ev- availability, an increase in luminal pH secondary to an
idence that there is intense and pathophysiologically relevant increase in ammonia concentration may be responsible
bidirectional crosstalk between microbiota and host; on the one for CKD-induced dysbiosis [16].
hand, uremia affects both the composition and metabolism of
the microbiota and, on the other hand, important uremic toxins Renal failure and microbial metabolism
originate uniquely from microbial metabolism. In addition, gut
dysbiosis may induce a disruption of the epithelial barrier, ulti- Poesen et al. recently compared the metabolite profiles of fecal
mately resulting in increased exposure of the host to endotoxins samples obtained from uremic and non-uremic patients and in
(lipopolysaccharides). These topics are discussed in more detail an animal model of uremia [19]. CKD was found to be asso-
in the following sections. ciated with a distinct colonic microbial metabolism, although
the effect of renal function loss per se in humans may be
inferior to the effects of dietary and other CKD-related factors.
Impact of renal failure on microbiota and microbial Impaired small intestinal protein assimilation [20], decreased
metabolism dietary fiber intake (due to reduced intake of fruit and vegeta-
bles, see following sections) [21] and slow colonic transit
Renal failure and microbiota (secondary to sedentary lifestyle, polypharmacia, restricted
intake of fluid, and uremia per se) [22] all foster protein fer-
Studies evaluating the gut bacterial flora in uremic patients mentation in CKD patients. The additional observation of
are scarce. Hida et al. studied uremic patients undergoing 24 h-urinary excretion of p-cresol paralleling renal function
Pediatr Nephrol

deterioration supports the notion of CKD being a state of metabolism and toxicity of these co-metabolites will be
increased protein fermentation [20]. discussed in more detail in subsections p-Cresyl sulfate and
indoxyl sulfate and Trimethylamine-N-oxide, respectively
(Table 1).
Impact of microbiota and microbial metabolism
on renal failure p-Cresyl sulfate and indoxyl sulfate

Leaky gut Metabolism p-Cresol is a putrefaction metabolite of tyrosine,


while indole is generated by the fermentation of tryptophan.
Chronic kidney disease is associated with increased intestinal After absorption, the majority of the p-cresol and indole is
(colonic) permeability (often referred to as ‘leaky gut’) [23]. further metabolized and conjugated to form p-cresyl sulfate
Gut dysbiosis may contribute to edema of the intestinal wall and indoxyl sulfate, respectively. Both p-cresyl sulfate and
and gut ischemia in patients with CKD, as well as to an im- indoxyl sulfate circulate tightly bound to albumin [34] and
paired colonic epithelial barrier [24]. A marked reduction of are cleared by the kidneys, mainly through tubular secretion.
the tight junction proteins claudin-1, occludin and zona The concentrations of p-cresyl sulfate and indoxyl sulfate in-
occludens-1 has been described in the gut mucosa of CKD crease concomitantly with the progression of CKD [35, 36], as
animals, probably due to the effects of uremic toxins [17, 24]. a consequence of both decreased elimination and increased
A leaky gut may represent an underappreciated cause of production [37], to reach concentrations in patients with
micro-inflammation [24, 25], a common condition in CKD end-stage renal disease (ESRD) that are 10- to 50-fold higher
and thought to contribute to malnutrition, accelerated cardio- than those in healthy controls. p-Cresyl sulfate and indoxyl
vascular disease and CKD progression [23, 26]. It remains to sulfate concentrations also increase with age [38]. Given their
be demonstrated whether persistent bacterial leakage of the strong albumin binding [39], multi-compartmental distribu-
gut triggering the phenomenon of ‘endotoxin tolerance’ may tion [40] and dependence on tubular secretion, the elimination
explain the concomitant acquired immunodeficiency of CKD of p-cresyl sulfate and indoxyl sulfate by conventional HD is
patients [23]. limited [41]. Following successful renal transplantation, the
Experimental data suggest that dietary fiber may reverse concentrations of these metabolites rapidly drop below levels
gut dysbiosis and suppress micro-inflammation [27]. These encountered in CKD counterparts matched for estimated glo-
data are in agreement with epidemiological evidence showing merular filtration rate (eGFR) [42].
associations between higher dietary fiber intake, better kidney
function [28] and lower inflammation, at least in the general Toxicity The toxicity of p-cresyl sulfite and indoxyl sulfate
population [28, 29]. has been demonstrated in numerous experimental studies [43]:
indoxyl sulfate and/or p-cresyl sulfate induce insulin resis-
Uremic toxins originating from microbial metabolism tance [44], suppress erythropoiesis [45], suppress klotho
[46–49] and trigger premature cellular senescence [50], acti-
Colonic microbiota are increasingly acknowledged to be an vate the renin–angiotensin–aldosterone system [51], cause en-
important source of uremic toxins. Aronov et al. compared the dothelial dysfunction [52, 53] and promote vascular calcifica-
plasma metabolite profile of HD patients with and without tion [54, 55], atherogenesis [56], thrombosis [57–59] and left
colons. Substantial amounts of p-cresyl sulfate and indoxyl ventricular hypertrophy and diastolic dysfunction [60]. They
sulfate were only observed in patients with an intact colon also induce renal tubulo-interstial fibrosis [61]. The toxic ef-
[30]. These findings extend data from a seminal animal study fects of p-cresyl sulfate and indoxyl sulfate are mediated at
by Wikoff et al. who compared the plasma from germ-free least in part by increased intracellular oxidative stress and
mice with that of wild-type mice by means of untargeted activation of the MAP kinase signaling pathway. Indoxyl sul-
metabolomics analyses [31]. These authors were able to iden- fate may also exert its toxic effects through activating the aryl
tify over 150 compounds, including p-cresyl sulfate and in- hydrocarbon signaling pathway [45, 55, 59, 62]. Clinical stud-
doxyl sulfate, that were unique to the wild-type mice. Another ies have consistently demonstrated (independent) associations
means to prove the microbial origin of a compound is to dem- between these metabolites and renal disease progression, car-
onstrate its plasma disappearance under broad spectrum anti- diovascular morbidity and mortality in patients with varying
biotic therapy, as was demonstrated for trimethylamine-N-ox- degrees of renal dysfunction [4, 35, 36, 63–67].
ide (TMAO) [32]. The list of uremic toxins, including those
originating from microbial metabolism, is expanding, mainly Trimethylamine-N-oxide
as a reflection of improved analytical techniques [33]. p-
Cresyl sulfate, indoxyl sulfate and TMAO are currently Metabolism Trimethylamine-N-oxide is an end-product of
attracting much attention as renovascular toxins. The microbial choline and carnitine metabolism [68, 69]. As a first
Pediatr Nephrol

Table 1 Phenols, indoles and trimethylamine-N-oxide: pathophysiology and treatment options

Pathophysiology/ Toxin Phenols (p-cresyl sulfate, phenyl Indoles (indoxyl sulfate, indoxyl Trimethylamine-N-oxide
treatment options acetyl glutamine, …) glucuronide, ..)

Pathophysiology Origin/source Tyrosine, phenylalanine Tryptophan Choline, carnitine


Impact of CKD Retention and increased production Retention and increased production Retention causes levels to
cause levels to increase up to cause levels to increase up to increase up to 30-fold
10-fold 10-fold
Toxicity Endothelial dysfunction, vascular Endothelial dysfunction, vascular Atherosclerosis, thrombosis,
calcification, thrombosis, atrial calcification, thrombosis, atrial renal fibrosis
fibrillation, renal fibrosis fibrillation, renal fibrosis
Treatment options Diet Low protein–high fiber diet Low protein–high fiber diet Mediterranean diet associates
associates with low toxin associates with low toxin with low TMAO exposure
exposure exposure
Prebiotic–probiotic– Pilot intervention studies show Pilot studies show promising, but No data
synbiotic promising, but not always not always consistent findings
consistent findings
Adsorbents No data Large RCT failed to confirm No data
promising results seen in
experimental and pilot clinical
intervention studies

CKD, Chronic kidney disease; TMAO, trimethylamine-N-oxide; RCT, randomized controlled trial

step, trimethylamine is transported via the portal circulation to at the Mayo Clinic, isolated a loop of ileum and by repeated
a cluster of hepatic enzymes, the flavin-containing perfusions with a lactated saline solution was able to prolong
monooxygenases, that efficiently oxidize it, thus forming the life of otherwise fatally ill young individuals with chronic
TMAO. TMAO enters the circulation, where it is predomi- uremia for more than 1 year [74]. As early as in 1951, Kolff
nantly excreted by the kidneys [70]. Concentrations of TMAO had acknowledged the large intestine as an import source/
increase concomitantly with the progression of CKD to reach reservoir of toxins and explored the therapeutic potential of
concentrations in patients with ESRD that are approximately intestinal lavage and oral sorbents [75]. Although with the
20-fold higher than those in healthy controls [71, 72]. TMAO advent of dialysis, renal transplantation and potent drug ther-
concentrations rapidly decline after successful renal transplan- apy, interest in dietary and gastrointestinal interventions has
tation [71, 72] toward concentrations observed in CKD coun- waned [76], it must be recognized that patients on renal re-
terparts matched for eGFR [42]. placement therapy do continue to suffer from the effects of
disease and face a mortality risk that greatly exceeds that of
Toxicity Trimethylamine-N-oxide may enhance the accu- the general population. This condition is referred to as the
mulation of macrophage cholesterol and the development residual syndrome, and there is renewed interest in dietary
of atherosclerosis [69], increase platelet hyperreactivity and gastrointestinal interventions as adjuvant therapies.
and thrombosis potential [73] and promote progressive
renal tubulo-interstitial fibrosis [70]. TMAO is increasing- Dietary interventions
ly being recognized as a predictor of cardiovascular dis-
ease, both in the general population [32, 68] and in pa- Dietary interventions and nutriceuticals are a logical co-
tients with CKD [71, 72]. A clinical link with progressive strategy to re-establish microbial balance and suppress circu-
renal disease is lacking as yet. lating p-cresyl sulfate, indoxyl sulfate and TMAO levels.
Dietary measures to restrict the intake of food rich in
(phosphatidyl)choline or carnitine may be helpful to lower
Interventions targeting toxic co-metabolites the exposure to TMAO. When protein fermentation metabo-
lites are the therapeutic target, both dietary protein restriction
Dietary and gastrointestinal approaches targeting colonic and dietary fiber supplementation may be considered.
co-metabolism Limiting dietary protein, although efficacious in limiting pro-
tein fermentation, as explained earlier in this review, may be
Dietary and gastrointestinal interventions have long been a ill-advised because of associated risks of protein malnutrition,
cornerstone of medicine. In the middle of the twentieth cen- especially in susceptible populations. A valuable and safe al-
tury, induced diarrhea and intestinal perfusion were popular ternative to dietary protein restriction to suppress protein fer-
treatments for advanced uremia. In 1990, Schloerb, a surgeon mentation is dietary fiber supplementation, as also explained
Pediatr Nephrol

earlier. Total dietary fiber intake reported in the third National demonstrated in numerous preclinical studies. Clinical studies
Health and Nutrition Examination Survey (NHANES III) av- have been less convincing. A phase 2 dose-finding study in
eraged 17 g/day, which is far below the 20–30 g/day recom- U.S. patients with CKD confirmed a dose-dependent reduc-
mended by current guidelines for the general population. tion in indoxyl sulfate serum concentrations [84]. Two rela-
Dietary fiber intake was even less in patients with CKD tively small and uncontrolled trials in Japanese patients with
(15 g/day). This observation is not unexpected as food items mild to moderate CKD [88, 89] suggested that AST-120 re-
rich in dietary fiber, such as fruits, vegetables and whole wheat tards the progression of renal failure. The multinational, ran-
bread, are often restricted in patients with advanced CKD to domized, double-blind, placebo-controlled Evaluating
prevent or correct hyperkalemia [77] and hyperphosphatemia. Prevention of Progression in CKD (EPPIC)-1 and EPPIC-2
In these patients, non-digestible oligo- and polysaccharides trials evaluated the effects of AST-120 on the progression of
[such as inulin, its hydrolysis product oligofructose and CKD when added to standard therapy [90]. More than 2000
(trans)galacto-oligosaccharides], often referred to as prebi- patients with moderate to severe CKD were randomized to
otics, may be considered to be a valuable and safe alternative receive either placebo or AST-120 (9 g/day). Unfortunately,
source of dietary fiber. Prebiotics are generally defined as the study failed to match expectations. Time to primary end-
selectively fermented ingredients that result in specific chang- point, i.e. a composite of dialysis initiation, kidney transplan-
es in the composition and/or activity of the gastrointestinal tation and serum creatinine doubling, was similar between the
microbiota (i.e. mainly bifidobacteria), thus conferring bene- AST-120 and the placebo groups in both trials. The study,
fit(s) upon host health [78]. In a pilot study in HD patients however, was hampered by several setbacks. Despite the sig-
(n = 22, uncontrolled), Meijers et al. demonstrated a signifi- nificant number of study participants, disease progression was
cant reduction in serum p-cresyl sulfate but not indoxyl sulfate more gradual than anticipated, and fewer patients reached the
following intake of oligofructose-enriched inulin [79]. In an- primary end-point, thus reducing statistical power. In addition,
other study in HD patients (n = 56, placebo-controlled), Sirich non-adherence to the study drug, related to the large pill bur-
et al. noted that the consumption of resistant starch significant- den, was also significant.
ly decreased serum indoxyl sulfate and, possibly, p-cresyl sul- Sevelamer hydrochloride (Renagel®; Genzyme, Boston,
fate [80]. One small study also investigated the effect of inulin MA), a non-calcium-containing phosphate binder, has been
in patients with CKD not yet on dialysis (n = 13, placebo- demonstrated in in vitro studies to also bind indole (10–
controlled cross-over) [81], demonstrating a significant de- 15 %) and p-cresol (40–50 %, pH-dependent) [91]. Despite
crease in serum p-cresol/p-cresyl sulfate, while the effect on this encouraging observation, however, it did not result in
other microbial metabolites was not studied; in contrast, there decreased serum concentrations of indoxyl sulfate or p-cresyl
was no effect of prebiotic arabinoxylan oligosaccharides sulfate either in a mouse model of CKD [92], or in a clinical
(n = 40, placebo-controlled cross-over) on solute levels of p- cross-over study. On the contrary, in the latter study there was
cresyl sulfate, indoxyl sulfate, TMAO and various other mi- a significant rise in serum p-cresyl sulfate during sevelamer
crobial metabolites in patients with an eGFR of between 15 treatment [93].
and 45 ml/min/1.73 m2. Therefore, further research remains
necessary to elucidate the role of prebiotic therapy in CKD. Drugs interfering with microbial or human metabolism
Of interest, vegetarians and vegans have lower circulating
levels of TMAO, p-cresyl sulfate and indoxyl sulfate than Probiotics [17, 94] and antimicrobials may also prove benefi-
omnivores [68, 82], probably due to a concomitant higher cial, but efficacy may be short-lasting. Probiotics have been
intake of dietary fiber and lower intake of protein. defined as Bviable organisms that, when ingested in sufficient
amounts, exert positive health effects^. Although numerous
Adsorbents studies have evaluated the effects of probiotics, only a limited
number have looked at their effects in renal disease.
Another approach is to reduce the availability of generated Moreover, these studies only assessed biochemical end-
microbial metabolites by adsorption of these metabolites onto points [76]. Preliminary data with host and bacterial enzyme
high-affinity surfaces. AST-120 (Kremezin®; Kureha inhibitors are promising [10, 95] and call for additional
Chemical Industry Co, Ltd., Tokyo, Japan) is an orally admin- studies.
istered adsorbent consisting of spherical carbon particles with
a diameter of 0.2–0.4 mm [83]. It is capable of adsorbing
significant amounts of various organic compounds in the large Future directions and perspectives
intestine, including indole [84, 85], p-cresol [86] and food-
derived advanced glycation end-products [87]. The efficacy It has long been known that the kidneys secrete organic acids
of AST-120 in decreasing circulating indoxyl sulfate levels originating from colonic microbial metabolism. However,
and preventing renal, vascular and bone damage has been study of the complex bidirectional interactions between the
Pediatr Nephrol

gut and the kidneys is still in its infancy and knowledge re- Compliance with ethical standards
mains limited. A better understanding of the gut–kidney axis
Conflict of interest The authors declare that they have no conflict of
will undoubtedly improve the care of patients with CKD and
interest.
progressive renal disease.
Growing evidence indicates that gut dysbiosis confers
health risks. A key question is how to define eubiosis. Valid
and at the same time workable biomarkers of gut eu-/dysbiosis
need to be identified. This is a challenging quest given the References
complexity of the microbial ecosystem. It is unlikely that a
single co-metabolite or germ is the culprit of all evil. 1. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W,
Biological variability, moreover, will prove to be high. In ad- Pettersson S (2012) host–gut microbiota metabolic interactions.
dition, the pathophysiological mechanisms underlying gut Science 336:1262–1267
2. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R
dysbiosis in CKD remain largely obscure. A better knowledge (2012) Diversity, stability and resilience of the human gut microbi-
of these mechanisms may help to halt the vicious circle finally ota. Nature 489:220–230
leading to ESRD and inherent comorbidity. 3. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh
Gut dysbiosis in CKD can be targeted in several ways. SA, Bewtra M, Knights D, Walters WA, Knight R, Sinha R, Gilroy
E, Gupta K, Baldassano R, Nessel L, Li H, Bushman FD, Lewis JD
From a teleological point of view, nutritional therapy is the
(2011) Linking long-term dietary patterns with gut microbial
most appealing. Pilot intervention trials with ‘nutriceuticals’ enterotypes. Science 334:105–108
have so far assessed biochemical outcomes and, overall, 4. Evenepoel P, Meijers BKI, Bammens BRM, Verbeke K (2009)
yielded promising results. Treatment response, however, was Uremic toxins originating from colonic microbial metabolism.
not always consistent, nor significant, possibly due to statisti- Kidney Int 76:S12–S19
5. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE,
cal issues and shortcomings of the intervention. Once the op- Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA,
timal treatment regimen has been defined, its efficacy needs to Biddinger SB, Dutton RJ, Turnbaugh PJ (2014) Diet rapidly and
be tested in adequately powered randomized controlled hard reproducibly alters the human gut microbiome. Nature 505:
end-point trials as this is the only way to get final proof of 559–563
6. Graf D, Di CR, Fak F, Flint HJ, Nyman M, Saarela M, Watzl B
concept. (2015) Contribution of diet to the composition of the human gut
Pharmaceuticals may have unexpected side effects on gut microbiota. Microb Ecol Health Dis 26:26164
microbiome and metabolism. This may apply to drugs com- 7. Zoetendal EG, de Vos WM (2014) Effect of diet on the intestinal
monly used in the care of renal patients, such as phosphate microbiota and its activity. Curr Opin Gastroenterol 30:189–195
8. Evenepoel P, Claus D, Geypens B, Hiele M, Geboes K, Rutgeerts P,
binders [93], proton pump inhibitors [96] and immunosup- Ghoos Y (1999) Amount and fate of egg protein escaping assimi-
pressants, among others. Researchers and clinicians should lation in the small intestine of humans. Am J Physiol Gastrointest
be aware of potential unwanted effects of drug therapy on Liver Physiol 277:G935–G943
the gut ecosystem. More efforts should be done to inventory 9. Macia L, Tan J, Vieira AT, Leach K, Stanley D, Luong S, Maruya
M, Ian MC, Hijikata A, Wong C, Binge L, Thorburn AN, Chevalier
these effects.
N, Ang C, Marino E, Robert R, Offermanns S, Teixeira MM,
Moore RJ, Flavell RA, Fagarasan S, Mackay CR (2015)
Metabolite-sensing receptors GPR43 and GPR109A facilitate die-
Conclusion tary fibre-induced gut homeostasis through regulation of the
inflammasome. Nat Commun 6:6734
10. Brown JM, Hazen SL (2015) The gut microbial endocrine organ:
It is increasingly acknowledged that crosstalk between host bacterially derived signals driving cardiometabolic diseases. Annu
and endogenous microbiota is pathophysiologically relevant Rev Med 66:343–359
in patients with CKD. The interactions are bidirectional; on 11. Cummings JH, Hill MJ, Bone ES, Branch WJ, Jenkins DJ (1979)
the one hand, uremia affects both the composition and metab- The effect of meat protein and dietary fiber on colonic function and
metabolism. II. Bacterial metabolites in feces and urine. Am J Clin
olism of the gut microbiota and, on the other hand, dysbiosis is Nutr 32:2094–2101
associated with an increased generation of microbiota-derived 12. Gabriele S, Sacco R, Altieri L, Neri C, Urbani A, Bravaccio C,
uremic toxins and micro-inflammation, both promoting the Riccio MP, Iovene MR, Bombace F, De ML, Persico AM (2016)
progression of renal disease and premature cardiovascular dis- Slow intestinal transit contributes to elevate urinary p-cresol level in
Italian autistic children. Autism Res 9:752–759
ease. Knowledge is rapidly accruing, but remains fragmentary. 13. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R,
Additional efforts are needed to unravel factors modulating Gordon JI (2007) The human microbiome project. Nature 449:
microbiota composition and microbial metabolism as well as 804–810
the underlying pathophysiological mechanisms. A better 14. Hida M, Aiba Y, Sawamura S, Suzuki N, Satoh T, Koga Y (1996)
Inhibition of the accumulation of uremic toxins in the blood and
knowledge of the impact of the kidneys on the gut microbiota their precursors in the feces after oral administration of Lebenin, a
ecosystem and vice versa is important as it may open new lactic acid bacteria preparation, to uremic patients undergoing he-
avenues for preventive therapy. modialysis. Nephron 74:349–355
Pediatr Nephrol

15. Wang IK, Lai HC, Yu CJ, Liang CC, Chang CT, Kuo HL, Yang YF, 32. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y,
Lin CC, Lin HH, Liu YL, Chang YC, Wu YY, Chen CH, Li CY, Hazen SL (2013) Intestinal microbial metabolism of phosphatidyl-
Chuang FR, Huang CC, Lin CH, Lin HC (2012) Real-time PCR choline and cardiovascular risk. N Engl J Med 368:1575–1584
analysis of the intestinal microbiotas in peritoneal dialysis patients. 33. Tanaka H, Sirich TL, Plummer NS, Weaver DS, Meyer TW (2015)
Appl Environ Microbiol 78:1107–1112 An enlarged profile of uremic solutes. PLoS One 10:e0135657
16. Vaziri ND, Wong J, Pahl M, Piceno YM, Yuan J, Desantis TZ, Ni Z, 34. Meijers BKI, Bammens B, Verbeke K, Evenepoel P (2008) A re-
Nguyen TH, Andersen GL (2013) Chronic kidney disease alters view of albumin binding in CKD. Am J Kidney Dis 51:839–850
intestinal microbial flora. Kidney Int 83:308–315 35. Meijers BKI, Claes K, Bammens B, de Loor H, Viaene L, Verbeke
17. Yoshifuji A, Wakino S, Irie J, Tajima T, Hasegawa K, Kanda T, K, Kuypers D, Vanrenterghem Y, Evenepoel P (2010) p-Cresol and
Tokuyama H, Hayashi K, Itoh H (2016) Gut Lactobacillus protects cardiovascular risk in mild-to-moderate kidney disease. Clin J Am
against the progression of renal damage by modulating the gut Soc Nephrol 5:1182–1189
environment in rats. Nephrol Dial Transplant 31:401–412 36. Barreto FC, Barreto DV, Liabeuf S, Meert N, Glorieux G, Temmar
18. Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri M, Choukroun G, Vanholder R, Massy ZA (2009) Serum indoxyl
ND (2014) Expansion of urease- and uricase-containing, indole- sulphate is associated with vascular disease and mortality in chronic
and p-cresol-forming and contraction of short-chain fatty acid- kidney disease patients. Clin J Am Soc Nephrol 4:1551–1558
producing intestinal microbiota in ESRD. Am J Nephrol 39: 37. Poesen R, Viaene L, Verbeke K, Claes K, Bammens B, Sprangers
230–237 B, Naesens M, Vanrenterghem Y, Kuypers D, Evenepoel P, Meijers
19. Poesen R, Windey K, Neven E, Kuypers D, De Preter V, Augustijns B (2013) Renal clearance and intestinal generation of p-cresyl sul-
P, D’Haese P, Evenepoel P, Verbeke K, Meijers B (2016) The in- phate and indoxyl sulphate in CKD. Clin J Am Soc Nephrol 8:
fluence of CKD on colonic microbial metabolism. J Am Soc 1508–1514
Nephrol 27:1389–1399 38. Viaene L, Thijs L, Jin Y, Liu Y, Gu Y, Meijers B, Claes K, Staessen
20. Bammens B, Verbeke K, Vanrenterghem Y, Evenepoel P (2003) J, Evenepoel P (2014) Heritability and clinical determinants of se-
Evidence for impaired assimilation of protein in chronic renal fail- rum indoxyl sulphate and p-cresyl sulphate, candidate biomarkers
ure. Kidney Int 64:2196–2203 of the human microbiome enterotype. PLoS One 9:e79682
21. Kalantar-Zadeh K, Kopple J, Deepak S, Block D, Block G (2002) 39. Viaene L, Annaert P, de Loor H, Poesen R, Evenepoel P, Meijers B
Food intake characteristics of hemodialysis patients as obtained by (2013) Albumin is the main plasma binding protein for indoxyl
food frequency questionnaire. J Ren Nutr 12:17–31 sulphate and p-cresyl sulphate. Biopharm Drug Dispos 34:165–175
22. Wu MJ, Chang CS, Cheng CH, Chen CH, Lee WC, Hsu YH, Shu 40. Cornelis T, Eloot S, Vanholder R, Glorieux G, van der Sande FM,
KH, Tang MJ (2004) Colonic transit time in long-term dialysis Scheijen JL, Leunissen KM, Kooman JP, Schalkwijk CG (2015)
patients. Am J Kidney Dis 44:322–327 Protein-bound uraemic toxins, dicarbonyl stress and advanced
23. Anders HJ, Andersen K, Stecher B (2013) The intestinal microbi- glycation end products in conventional and extended haemodialysis
ota, a leaky gut, and abnormal immunity in kidney disease. Kidney and haemodiafiltration. Nephrol Dial Transplant 30:1395–1402
Int 83:1010–1016 41. Meyer TW, Peattie JW, Miller JD, Dinh DC, Recht NS, Walther JL,
24. Vaziri ND (2012) CKD impairs barrier function and alters microbial Hostetter TH (2007) Increasing the clearance of protein-bound sol-
flora of the intestine: a major link to inflammation and uremic utes by addition of a sorbent to the dialysate. J Am Soc Nephrol 18:
toxicity. Curr Opin Nephrol Hypertens 21:587–592 867–874
25. Poesen R, Ramezani A, Claes K, Augustijns P, Kuypers D, Barrows 42. Poesen R, Evenepoel P, de Loor H, Bammens B, Claes K,
IR, Muralidharan J, Evenepoel P, Meijers B, Raj DS (2015) Sprangers B, Naesens M, Kuypers D, Augustijns P, Meijers B
Associations of soluble CD14 and endotoxin with mortality, cardio- (2016) The influence of renal transplantation on retained
vascular disease, and progression of kidney disease among patients microbial-human co-metabolites. Nephrol Dial Transplant 31:
with CKD. Clin J Am Soc Nephrol 10:1525–1533 1721–1729
26. Stenvinkel P, Heimburger O, Paultre F, Diczfalusy U, Wang T, 43. Vanholder R, Schepers E, Pletinck A, Nagler EV, Glorieux G
Berglund L, Jogestrand T (1999) Strong association between mal- (2014) The uremic toxicity of indoxyl sulphate and p-cresyl sul-
nutrition, inflammation, and atherosclerosis in chronic renal failure. phate: a systematic review. J Am Soc Nephrol 25:1897–1907
Kidney Int 55:1899–1911 44. Koppe L, Pillon NJ, Vella RE, Croze ML, Pelletier CC, Chambert
27. Vaziri ND, Liu SM, Lau WL, Khazaeli M, Nazertehrani S, S, Massy Z, Glorieux G, Vanholder R, Dugenet Y, Soula HA,
Farzaneh SH, Kieffer DA, Adams SH, Martin RJ (2014) High Fouque D, Soulage CO (2013) p-Cresyl sulphate promotes insulin
amylose resistant starch diet ameliorates oxidative stress, inflam- resistance associated with CKD. J Am Soc Nephrol 24:88–99
mation, and progression of chronic kidney disease. PLoS One 9: 45. Asai H, Hirata J, Hirano A, Hirai K, Seki S, Watanabe-Akanuma M
e114881 (2016) Activation of aryl hydrocarbon receptor mediates suppres-
28. Xu H, Huang X, Riserus U, Krishnamurthy VM, Cederholm T, sion of hypoxia-inducible factor-dependent erythropoietin expres-
Arnlov J, Lindholm B, Sjogren P, Carrero JJ (2014) Dietary fiber, sion by indoxyl sulphate. Am J Physiol Cell Physiol 310:
kidney function, inflammation, and mortality risk. Clin J Am Soc C142–C150
Nephrol 9:2104–2110 46. Shimizu H, Bolati D, Adijiang A, Adelibieke Y, Muteliefu G,
29. Krishnamurthy VM, Wei G, Baird BC, Murtaugh M, Chonchol Enomoto A, Higashiyama Y, Higuchi Y, Nishijima F, Niwa T
MB, Raphael KL, Greene T, Beddhu S (2012) High dietary fiber (2011) Indoxyl sulphate downregulates renal expression of Klotho
intake is associated with decreased inflammation and all-cause mor- through production of ROS and activation of nuclear factor-kB. Am
tality in patients with chronic kidney disease. Kidney Int 81: J Nephrol 33:319–324
300–306 47. Adijiang A, Shimizu H, Higuchi Y, Nishijima F, Niwa T (2011)
30. Aronov PA, Luo FJ, Plummer NS, Quan Z, Holmes S, Hostetter Indoxyl sulphate reduces klotho expression and promotes senes-
TH, Meyer TW (2011) Colonic contribution to uremic solutes. J cence in the kidneys of hypertensive rats. J Ren Nutr 21:105–109
Am Soc Nephrol 22:1769–1776 48. Adijiang A, Niwa T (2010) An oral sorbent, AST-120, increases
31. Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Klotho expression and inhibits cell senescence in the kidney of
Siuzdak G (2009) Metabolomics analysis reveals large effects of uremic rats. Am J Nephrol 31:160–164
gut microflora on mammalian blood metabolites. Proc Natl Acad 49. Sun CY, Chang SC, Wu MS (2012) Suppression of Klotho expres-
Sci USA 106:3698–3703 sion by protein-bound uremic toxins is associated with increased
Pediatr Nephrol

DNA methyltransferase expression and DNA hypermethylation. 65. Liabeuf S, Barreto DV, Barreto FC, Meert N, Glorieux G, Schepers
Kidney Int 81:640–650 E, Temmar M, Choukroun G, Vanholder R, Massy ZA, on behalf of
50. Muteliefu G, Shimizu H, Enomoto A, Nishijima F, Takahashi M, the European Uraemic Toxin Work Group (EUTox) (2010) Free p-
Niwa T (2012) Indoxyl sulphate promotes vascular smooth muscle cresylsulphate is a predictor of mortality in patients at different
cell senescence with upregulation of p53, p21, and prelamin A stages of chronic kidney disease. Nephrol Dial Transplant 25:
through oxidative stress. Am J Physiol Cell Physiol 303: 1183–1191
C126–C134 66. Niewczas MA, Sirich TL, Mathew AV, Skupien J, Mohney RP,
51. Sun CY, Chang SC, Wu MS (2012) Uremic toxins induce kidney Warram JH, Smiles A, Huang X, Walker W, Byun J, Karoly ED,
fibrosis by activating intrarenal renin-angiotensin-aldosterone sys- Kensicki EM, Berry GT, Bonventre JV, Pennathur S, Meyer TW,
tem associated epithelial-to-mesenchymal transition. PLoS One 7: Krolewski AS (2014) Uremic solutes and risk of end-stage renal
e34026 disease in type 2 diabetes: metabolomic study. Kidney Int 85:
52. Meijers BK, Van KS, Verbeke K, Dehaen W, Vanrenterghem Y, 1214–1224
Hoylaerts MF, Evenepoel P (2009) The uremic retention solute p- 67. Lin CJ, Wu V, Wu PC, Wu CJ (2015) Meta-analysis of the associ-
cresyl sulphate and markers of endothelial damage. Am J Kidney ations of p-cresyl sulphate (P-CRESYL SULPHATE) and indoxyl
Dis 54:891–901 sulphate (IS) with cardiovascular events and all-cause mortality in
53. Six I, Gross P, Remond MC, Chillon JM, Poirot S, Drueke TB, patients with chronic renal failure. PLoS One 10:e0132589
Massy ZA (2015) Deleterious vascular effects of indoxyl sulphate 68. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt
and reversal by oral adsorbent AST-120. Atherosclerosis 243: EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu
248–256 GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH,
54. Niwa T, Shimizu H (2012) Indoxyl sulphate induces Bushman FD, Lusis AJ, Hazen SL (2013) Intestinal microbiota
nephrovascular senescence. J Ren Nutr 22:102–106 metabolism of L-carnitine, a nutrient in red meat, promotes athero-
55. Ochi A, Mori K, Nakatani S, Emoto M, Morioka T, Motoyama K, sclerosis. Nat Med 19:576–585
Fukumoto S, Imanishi Y, Shoji T, Ishimura E, Inaba M (2015) 69. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B,
Indoxyl sulphate suppresses hepatic fetuin-A expression via the aryl Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith
hydrocarbon receptor in HepG2 cells. Nephrol Dial Transplant 30: JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL (2011)
1683–1692 Gut flora metabolism of phosphatidylcholine promotes cardiovas-
56. Jing YJ, Ni JW, Ding FH, Fang YH, Wang XQ, Wang HB, Chen cular disease. Nature 472:57–63
XN, Chen N, Zhan WW, Lu L, Zhang RY (2016) p-Cresyl sulphate 70. Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle
is associated with carotid arteriosclerosis in hemodialysis patients B, Li XS, Levison BS, Hazen SL (2015) Gut microbiota-dependent
and promotes atherogenesis in apoE−/− mice. Kidney Int 89: trimethylamine N-oxide (TMAO) pathway contributes to both de-
439–449 velopment of renal insufficiency and mortality risk in chronic kid-
57. Wu CC, Hsieh MY, Hung SC, Kuo KL, Tsai TH, Lai CL, Chen JW, ney disease. Circ Res 116:448–455
Lin SJ, Huang PH, Tarng DC (2016) Serum indoxyl sulphate asso- 71. Stubbs JR, House JA, Ocque AJ, Zhang S, Johnson C, Kimber C,
ciates with postangioplasty thrombosis of dialysis grafts. J Am Soc Schmidt K, Gupta A, Wetmore JB, Nolin TD, Spertus JA, Yu AS
Nephrol 27:1254–1264 (2016) Serum trimethylamine-N-oxide is elevated in CKD and cor-
58. Chitalia VC, Shivanna S, Martorell J, Balcells M, Bosch I, relates with coronary atherosclerosis burden. J Am Soc Nephrol 27:
Kolandaivelu K, Edelman ER (2013) Uremic serum and solutes 305–313
increase post-vascular interventional thrombotic risk through al- 72. Missailidis C, Hallqvist J, Qureshi AR, Barany P, Heimburger O,
tered stability of smooth muscle cell tissue factor. Circulation Lindholm B, Stenvinkel P, Bergman P (2016) Serum
127:365–376 trimethylamine-N-oxide is strongly related to renal function and
59. Shivanna S, Kolandaivelu K, Shashar M, Belghasim M, Al-Rabadi predicts outcome in chronic kidney disease. PLoS One 11:
L, Balcells M, Zhang A, Weinberg J, Francis J, Pollastri MP, e0141738
Edelman ER, Sherr DH, Chitalia VC (2016) The aryl hydrocarbon 73. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu
receptor is a critical regulator of tissue factor stability and an anti- X, Wu Y, Mehrabian M, Sartor RB, McIntyre TM, Silverstein RL,
thrombotic target in uremia. J Am Soc Nephrol 27:189–201 Tang WH, DiDonato JA, Brown JM, Lusis AJ, Hazen SL (2016)
60. Yang K, Wang C, Nie L, Zhao X, Gu J, Guan X, Wang S, Xiao T, Gut microbial metabolite TMAO enhances platelet hyperreactivity
Xu X, He T, Xia X, Wang J, Zhao J (2015) Klotho protects against and thrombosis risk. Cell 165:111–124
indoxyl sulphate-induced myocardial hypertrophy. J Am Soc 74. Schloerb PR (1990) Intestinal dialysis for kidney failure. Personal
Nephrol 26:2434–2446 experience. ASAIO Trans 36:4–7
61. Sun CY, Hsu HH, Wu MS (2013) p-Cresol sulphate and indoxyl 75. Twiss EE, Kolff WJ (1951) Treatment of uremia by perfusion of an
sulphate induce similar cellular inflammatory gene expressions in isolated intestinal loop; survival for forty-six days after removal of
cultured proximal renal tubular cells. Nephrol Dial Transplant 28: the only functioning kidney. J Am Med Assoc 146:1019–1022
70–78 76. Poesen R, Meijers B, Evenepoel P (2013) The colon: an overlooked
62. Schroeder JC, Dinatale BC, Murray IA, Flaveny CA, Liu Q, site for therapeutics in dialysis patients. Semin Dial 26:323–332
Laurenzana EM, Lin JM, Strom SC, Omiecinski CJ, Amin S, 77. Evenepoel P, Meijers BK (2012) Dietary fiber and protein: nutri-
Perdew GH (2010) The uremic toxin 3-indoxyl sulphate is a potent tional therapy in chronic kidney disease and beyond. Kidney Int 81:
endogenous agonist for the human aryl hydrocarbon receptor. 227–229
Biochemistry 49:393–400 78. Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall R,
63. Bammens B, Evenepoel P, Keuleers H, Verbeke K, Vanrenterghem Rowland I, Wolvers D, Watzl B, Szajewska H, Stahl B, Guarner F,
Y (2006) Free serum concentrations of the protein-bound retention Respondek F, Whelan K, Coxam V, Davicco MJ, Leotoing L,
solute p-cresol predict mortality in hemodialysis patients. Kidney Wittrant Y, Delzenne NM, Cani PD, Neyrinck AM, Meheust A
Int 69:1081–1087 (2010) Prebiotic effects: metabolic and health benefits. Br J Nutr
64. Meijers BKI, Bammens B, De Moor B, Verbeke K, Vanrenterghem 104[Suppl 2]:S1–S63
Y, Evenepoel P (2008) Free p-cresol is associated with cardiovas- 79. Meijers BK, De Preter V, Verbeke K, Vanrenterghem Y, Evenepoel
cular disease in hemodialysis patients. Kidney Int 73:1174–1180 P (2010) p-Cresyl sulphate serum concentrations in haemodialysis
Pediatr Nephrol

patients are reduced by the prebiotic oligofructose-enriched inulin. 89. Shoji T, Wada A, Inoue K, Hayashi D, Tomida K, Furumatsu Y,
Nephrol Dial Transplant 25:219–224 Kaneko T, Okada N, Fukuhara Y, Imai E, Tsubakihara Y (2007)
80. Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW Prospective randomized study evaluating the efficacy of the spher-
(2014) Effect of increasing dietary fiber on plasma levels of colon- ical adsorptive carbon AST-120 in chronic kidney disease patients
derived solutes in hemodialysis patients. Clin J Am Soc Nephrol 9: with moderate decrease in renal function. Nephron Clin Pract 105:
1603–1610 C99–C107
81. Salmean YA, Segal MS, Palii SP, Dahl WJ (2015) Fiber supple- 90. Schulman G, Berl T, Beck GJ, Remuzzi G, Ritz E, Arita K, Kato A,
mentation lowers plasma p-cresol in chronic kidney disease pa- Shimizu M (2015) Randomized placebo-controlled EPPIC Trials of
tients. J Ren Nutr 25:316–320 AST-120 in CKD. J Am Soc Nephrol 26:1732–1746
82. Patel KP, Luo FJ, Plummer NS, Hostetter TH, Meyer TW (2012) 91. de Smet R, Thermote F, Lameire N, Vanholder R (2004) Sevelamer
The production of p-cresol sulphate and indoxyl sulphate in vege- hydrochloride (Renagel) absorbs the uremic compounds indoxyl
tarians versus omnivores. Clin J Am Soc Nephrol 7:982–988 sulphate, indole and p-cresol [Abstract]. J Am Soc Nephrol 15:
83. Marier JF, Guilbaud R, Kambhampati SRP, Mathew P, Moberly J, 505A (abstract)
Lee J, Salazar DE (2006) The effect of AST-120 on the single-dose 92. Phan O, Ivanovski O, Nguyen-Khoa T, Mothu N, Angulo J,
pharmacokinetics of losartan and losartan acid (E-3174) in healthy Westenfeld R, Ketteler M, Meert N, Maizel J, Nikolov IG,
subjects. J Clin Pharmacol 46:310–320 Vanholder R, Lacour B, Drueke TB, Massy ZA (2005) Sevelamer
84. Schulman G, Agarwal R, Acharya M, Berl T, Blumenthal S, Kopyt prevents uremia-enhanced atherosclerosis progression in apolipo-
N (2006) A multicenter, randomized, double-blind, placebo-con- protein E-deficient mice. Circulation 112:2875–2882
trolled, dose-ranging study of AST-120 (Kremezin) in patients with 93. Brandenburg VM, Schlieper G, Heussen N, Holzmann S, Busch B,
moderate to severe CKD. Am J Kidney Dis 47:565–577 Evenepoel P, Vanholder R, Meijers B, Meert N, Fassbender WJ,
85. Niwa T, Nomura T, Sugiyama S, Miyazaki T, Tsukushi S, Tsutsui S Floege J, Jahnen-Dechent W, Ketteler M (2010) Serological cardio-
(1997) The protein metabolite hypothesis, a model for the progres- vascular and mortality risk predictors in dialysis patients receiving
sion of renal failure: an oral adsorbent lowers indoxyl sulphate sevelamer: a prospective study. Nephrol Dial Transplant 25:
levels in undialyzed uremic patients. Kidney Int 52:S23–S28 2672–2679
86. Niwa T, Ise M, Miyazaki T, Meada K (1993) Suppressive effect of 94. Koppe L, Mafra D, Fouque D (2015) Probiotics and chronic kidney
an oral sorbent on the accumulation of p-cresol in the serum of disease. Kidney Int 88:958–966
experimental uremic rats. Nephron 65:82–87 95. Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E,
87. Yamagishi S, Nakamura K, Matsui T, Inoue H, Takeuchi M (2007) Gu X, Huang Y, Zamanian-Daryoush M, Culley MK,
Oral administration of AST-120 (Kremezin) ia a promizing thera- DiDonato AJ, Fu X, Hazen JE, Krajcik D, DiDonato JA,
peutic strategy for advanced glycation end product (AGE)-related Lusis AJ, Hazen SL (2015) Non-lethal inhibition of gut micro-
disorders. Med Hypotheses 69:666–668 bial trimethylamine production for the treatment of atheroscle-
88. Owada K, Nakao M, Koike J, Ujiie K, Tomita K, Shiigai T (1997) rosis. Cell 163:1585–1595
Effects of oral adsorbent AST-120 on the progression of chronic 96. Poesen R, Meijers B, Evenepoel P (2016) Adverse effects of proton
renal failure: a randomized controlled study. Kidney Int 63: pump inhibitors in chronic kidney disease. JAMA Intern Med 176:
188–190 867–868

You might also like