Abstract
Also known as the “second human genome,” the gut microbiome plays important roles in both the maintenance of health and the pathogenesis of disease. The symbiotic relationship between host and microbiome is disturbed due to proliferation of dysbiotic bacteria in patients with chronic kidney disease (CKD). Fermentation of protein and amino acids by gut bacteria generate excess amounts of potentially toxic compounds such as ammonia, amines, thiols, phenols, and indoles, but generation of short chain fatty acids is reduced. Impaired intestinal barrier function in CKD permits translocation of gut-derived uremic toxins into the systemic circulation, contributing to progression of CKD, cardiovascular disease, insulin resistance, and protein energy wasting. The field of microbiome research is still nascent, but evolving rapidly. Establishing symbiosis to treat uremic syndrome is a novel concept, but if proven effective will have significant impact on the management of patients with CKD.
Index words: gut microbiome, uremic toxin, microbial metabolite, metabolome, ammonia, urea, amine, thiol, phenol, indole, p-cresol sulfate (PCS), uremic syndrome, chronic kidney disease (CKD), end-stage renal disease (ESRD), review
Keywords: Gut microbiome, chronic kidney disease, uremic toxins
BACKGROUND
Findings from the Human Microbiome Project (HMP) and the Metagenomics of the Human Intestinal Tract (Meta-HIT) project have shown that the human intestine is home to an extraordinarily complex and dynamic consortium of bacteria that play a pivotal role in human health and disease.1, 2 Bacteria have co-evolved with humans, and this symbiotic relationship has expanded our capabilities beyond what is coded in our own genome.3 Indeed, genetically, we are vastly outnumbered by our own microbiome, the microbial genome. As the Nobel Laureate Joshua Lederberg has asserted, “We should think of each host and its parasites as a superorganism with the respective genomes yoked into a chimera of sorts.”4 The central role of gut in human health has been long recognized, dating back to 400 B.C. when Hippocrates stated, “death sits in the bowels.”5 This review will provide an overview of the bidirectional relationship between chronic kidney disease (CKD) and the gut microbiome, discuss the consequence of gut dysbiosis in the pathogenesis of systemic inflammation and uremic toxicity, and highlight the recent advances in targeting gut microbiome for therapeutic purposes.
CASE VIGNETTE
A 65-year-old man with CKD stage G4 presented with lethargy and chronic constipation to the emergency room. Clinical examination was unremarkable except for generalized muscle weakness and distended abdomen with sluggish bowel sounds. Laboratory investigation showed a sodium level of 138 meq/L; potassium, 6.3 meq/L; chloride, 115 meg/L; bicarbonate, 16 meq/L; anion gap, 14; serum urea nitrogen, 60 mg/dl; serum creatinine, 3.8 mg/dl (correspondign to an estimated GFR of 16 ml/min/1.73 using the *** equation); glucose, 100 mg/dL; calcium, 8.1 mg/dL; phosphate, 7.1 mg/dL; albumin, 3.6 g/dL; WBC count, 8.1 × 109/L; and hemoglobin, 10.1 g/dL. Computed tomography (CT) of the brain was normal except for mild cortical atrophy. Abdominal CT showed abundant fecal matter in a dilated rectum and sigmoid colon. After disimpaction with enemas and laxatives, the patient felt better. He was discharged with recommendation to take laxatives on a regular basis.
He was seen in the outpatient clinic two months later. He appeared energetic and said that he was taking a prebiotic (p-inulin) and continuing the laxative when needed. Repeat laboratory evaluation showed a sodium level of 139 meq/L; potassium, 4.0 meq/L; chloride, 110 meq/L; bicarbonate, 20 meq/L; anion gap, 11; serum urea nitrogen, 51 mg/dl; serum creatinine, 3.4 mg/dL (correspondign to an eGFR of 18 ml/min/1.73 m2); glucose, 82 mg/dL; calcium, 8.3 mg/dL; phosphate, 6.2 mg/dL; and albumin, 3.8 g/dL.
In the case presented, the patient’s clinical symptoms and biochemistry improved with relief of constipation, possibly through decreased generation and increased elimination of uremic toxins. This highlights the importance of colon health in patients with CKD.
PATHOGENESIS
Gut microbiome in health
The human gut harbors ~1014 bacteria with an enormous metabolic potential.6–8 Under physiologic conditions, the microbiota provide complementary functions by participating in metabolic activities that are not fully evolved in the human host, such as digestion of complex polysaccharides,9 endogenous synthesis of certain vitamins and amino acids,10 metabolism of bile acids,11 degradation of dietary oxalates,12 and maturation of the immune system.13
On average, an individual’s gut microbiota is composed of 500–1,000 bacterial species.14 Findings from the HMP suggest that each individual has a unique microbiome, each niche features one or a few signature taxa, and the gut microbiome is characterized by the greatest diversity with little variation over time.15, 16 The predominant bacterial groups in the human gastrointestinal tract are Bacteroidetes, Firmicutes, and Actinobacteria.17, 18 The phylogenetic composition of gut microbiota tend to be similar between individuals living in the same region, belonging to the same family, and having a similar diet.19 Muegge et al. studied gut microbiome profile in 33 mammalian species, including 18 humans, and reported that the difference in microbiome profile stems from differing metabolic functions required to utilize the diet.20 Thus, the gut microbiome appears to change adaptively to the needs of the host organism.
Gut Microbiome in Kidney Disease
The term “dysbiosis” was first coined in early twentieth century by the Russian Nobel Laureate Elie Metchnikoff.21 Dysbiosis is defined as an imbalanced intestinal microbial community with quantitative and qualitative alterations in the composition and in metabolic activities of the gut microbiota. Preliminary evidence indicates that microbiome profile might be altered in patients with chronic kidney failure and earlier stages of CKD.22 (Table 1) Vaziri et al. found that 190 microbial operational taxonomic units (OTUs) differeed significantly in abundance between ESRD patients and apparently healthy controls.23 Hida et al. reported that the number of aerobic bacteria including Enterobacteria and Enterococci is higher in patients treated with maintenance hemodialysis than in controls.24 Among anaerobic bacteria, Hida et al observed that hemodialysis patients have significantly lower number of Bifidobacteria and higher organism counts for Clostridium perfringens.24
Table 1.
Alterations of gut microbiome in CKD
Study group | Methods | Gut microbiota features associated with CKD | Ref. |
---|---|---|---|
Human studies | |||
CKD patients (n=22), with (n=12) and without (n=10) GI symptoms | Antroduodenojejunal manometry, culture | Abnormal motility and bacterial overgrowth in small intestine of CKD patients | 153 |
Hemodialysis patients (n=8) | Bacterial cell count | Increased counts of both aerobic (~ 106 bacteria/ml) and anaerobic (~ 107 bacteria/ml) organisms in the duodenum and jejunum (which are normally not colonized heavily by bacteria in healthy individuals) | 154 |
ESRD patients (n=24) vs. healthy persons (n=12) | Phylogenetic microarray | Highly significant differences between the ESRD and the healthy control groups in the abundance of over 200 bacterial operational taxonomic units belonging to 23 bacterial families | 23 |
ESRD patients (n=24) vs. healthy persons (n=12) | Phylogenetic microarray and Hypergeometric distribution tests | Expansion of bacterial families possessing urease, uricase, and p-cresol- and indole-forming enzymes; depletion of bacteria possessing short-chain fatty acid forming enzymes | 155 |
ESRD patients (n=10) vs. healthy persons (n=8) | 16S rDNA amplification and DNA pyrosequencing | Disturbed composition of the microbiota characterized by an overgrowth of aerobic bacteria such as Enterobacteria and Enterococci species (~100 times higher in ESRD patients); of the anaerobic bacteria, ESRD patients had significantly lower numbers of Bifidobacteria and higher numbers of Clostridium perfringens | 24 |
ESRD patients (n=52) | 16S rDNA amplification and pyrosequencing | Increased bacterial translocation in plasma of ESRD patients; bacterial DNA concentration positively correlated with plasma levels of CRP and IL-6 | 156 |
Experimental animal studies | |||
5/6 nephrectomy (n=6) or sham- operated rats (n=5) | Phylogenetic microarray | Significant difference in the abundance of 175 bacterial operational taxonomic units between the uremic and control animals, most notably as decreases in the Lactobacillaceae and Prevotellaceae families | 23 |
Uremic (n=20) and control (n=20) Wistar rats | Culture | Impaired intestinal mucosa barrier with significantly more bacterial translocation in uremic than in control animals | 157 |
Abbreviation: CKD, chronic kidney disease; ESRD, end-stage renal disease; GI, gastrointestinal; rDNA, ribosomal DNA; CRP, C-reactive protein; IL, interleukin
The main contributing factors to gut microbiome dysbiosis in patients with kidney disease include slow intestinal transit time,25 impaired protein assimilation,26 decreased consumption of dietary fiber,27 iron therapy,28 and frequent use of antibiotics.29, 30 Antibiotic treatment decreases the diversity and alters the relative abundances of members of the bacterial community, with some patients exhibiting incomplete recovery post-treatment.31
Gut-Derived Uremic Toxins and Microbial Metabolites
In 1965, Einheber and Carter showed that germfree anephric mice survived longer than anephric mice with intact gut microbiome.32 Aronov et al. showed that a number of uremic retention solutes are present only in hemodialysis patients with intact colon.33 Recently, using untargeted metabolomic mass spectrometry, Wikoff et al. reported that the presence of several protein-bound uremic toxins, such as indoxyl sulfate (IS), hippuric acid, and phenylacetic acid are dependent on the presence of gut microflora.34
Impaired protein assimilation in uremia leads to large influx of undigested proteins into the distal intestine, which favors the proliferation of proteolytic bacteria.35 (Fig 1) Increased protein fermentation results in generation of potentially toxic metabolites, such as ammonia, phenols, amines, indoles, and thiols.36 The clinical manifestations of these uremic toxins are rather nonspecific and may include neurologic disorders, protein energy wasting, cardiovascular disease (CVD), and progression of CKD. The potential pathways linking the accumulation of some of the major toxic metabolites to patho-physiological consequences in patients with CKD are shown schematically in Fig 2. Increased levels of these toxins in CKD may be related to increased generation from dysbiotic microbiome or decreased elimination from reduced kidney function. In this review we will focus on the role of gut microbiome in generation of uremic toxins. (Table 2)
Figure 1.
Schematic representation of the association between uremia, dysbiotic gut microbiome, gut-derived uremic toxins, and the clinical manifestations of these uremic toxins.
Figure 2.
Schematic showing some of the major toxic metabolites originating from synthesis by dysbiotic gut microbiome and potential pathways linking their accumulations to pathophysiological consequences in CKD, including in ESRD. Increased intestinal concentration of uremic toxins associated with the progression of CKD leads to microbial dysbiosis. p-Cresol, produced by the intestinal microbiota from the amino acid tyrosine, inhibits colonocyte respiration and proliferation, and at higher concentrations, increases DNA damage and becomes genotoxic towards colonocytes.151 Overgrowth of pathogenic bacteria, the loss of barrier integrity, and the breach in the epithelia barrier lead to endotoxemia. Circulating endotoxin, also referred to as lipopolysaccharide (LPS), activates production of inflammatory cytokines. Endotoxin translocation from the gut has been suggested as one of the causes of inflammation in CKD.103, 113 Amongst the most studied uremic toxins are phospholipid metabolites and bacterial products of choline degradation, such as TMAO, which has been associated with CVD. TMAO causes alteration of cholesterol and sterol metabolism, promotes foam cell formation by increasing expression of scavenger receptors on macrophages, and leads to alternations in bile acid metabolism and sterol transporters both within the liver and intestine.103 Other uremic toxins include cometabolites of phenols (eg, PCS), and indoles (eg, IS), which have been associated with progression of CKD, CVD, and mortality in hemodialysis patients. Accumulation of PCS in human tubular cells leads to Nox4-dependent ROS generation via upregulation of Nox4 and p22phox, which subsequently enhance the expression of inflammatory cytokines and profibrotic factors, resulting in cell injury.152 Activation of PI3-K and PKC mediate the stimulatory effect of PCS on Nox4 and NADPH oxidase. IS induces nephrotoxicity via organic anion transporter–mediated uptake in the basolateral membrane of renal proximal tubular cells, where it activates NF-κB and plasminogen activator inhibitor type 1 expression.54 ROS, reactive oxygen species
Table 2.
Gut microbiome derived uremic toxins
Solute (MW) | Group | Source | Related bacteria | Pathogenies/Mechanism | Ref. |
---|---|---|---|---|---|
Free, water-soluble, low MW molecules (<0.5 kDa) | |||||
Ammonia (17 Da) | Bacterial hydrolysis of urea by urease ; bacterial fermentation of glutamine, serine, threonine and glycine | Urease is produced by diverse bacterial species; Clostridium spp., Enterococcus, Shigella and Escherichia coli | High concentration of ammonia changes the luminal pH causing uremic enterocolitis; amino acid catabolism leads to the formation of sulfides, phenolic compounds and amines, which are inflammatory and/or precursors to the formation of carcinogens | 158, 159 | |
1- Methyl guanidine (73 Da) | Guanidin e | Metabolism of creatinine | Pseudomonas stutzeri | Accumulate in CKD and considered a uremic toxin; administration to rats with kidney failure demonstrates a dose- dependent increase in mortality | 43–45 |
TMAO (75 Da) | Amine | Endogenous; bacterial metabolism of dietary lipid phosphatidylcholine | Faecalibacterium prausnitzii, Bifidobacterium | ↑ progression of kidney disease and mortality in CKD; ↑ in tubulointerstitial fibrosis and collagen deposition; ↑ phosphorylation of Smad3, which regulates the profibrotic TGFβ/Smad3 signaling | 160-162 |
Homocysteine (135 Da) | Amino acid | Endogenous; intestinal bacteria lower homocysteine by production of folic acid | Bifidobacterium spp. | ↑ CVD and death; ↑oxidative stress (through the production of reactive oxygen species), binds to nitric oxide, produces homocysteinylated proteins, and leads to the accumulation of its precursor, S-adenosylhomocysteine, a potent inhibitor of biological transmethylations | 163 |
D-lactic acid (90 Da) | D-amino acid | Ingestion; endogenous; bacterial production | Enterococcus and Streptococcus spp. | D-lactic acidosis; neurotoxic effects; encephalopathic symptoms | 100, 101 |
Oxalate (90 Da) | Ingestion; endogenous; certain intestinal bacteria have oxalate-degrading potency | Oxalobacter formigenes, B. lactis, E. faecalis, and Eubacterium lentum | Hyperoxaluria leading to urolithiasis; ↓ endothelial cell replication and migration leading to atherosclerosis | 164 | |
Protein-bound solutes | |||||
p-Cresol sulfate (188 Da) | Phenol | Bacterial metabolism of tyrosine and phenylalanine | Clostidium difficile, F. prausnitzii, Bifidobacterium, Subdoligranulum, Lactobacillus | ↑ progression of CKD, CVD, and mortality in hemodialysis patients; ↓ cytokine-stimulated expression of endothelial adhesion molecules; ↑ endothelial permeability | 78–81,165, 166 |
Indoxyl sulfate (213 Da) | Indole | Bacterial metabolism of tryptophan | Clostridium sporogenes, E. coli | ↑ vascular stiffness, aortic calcification, and cardiovascular mortality; ↑ oxidative stress in endothelial cells, ↑ vascular smooth muscle cell proliferation; ↑ expression of genes related to tubulointerstitial fibrosis; ↑ nephrotoxicity | 53, 55,56 |
Indole-3-acetic acid (175 Da) | Indole | Endogenous; bacterial metabolism of tryptophan | Clostridium sporogenes, C. bartlettii, E. coli | Induce glomerular sclerosis and interstitial fibrosis in subtotally nephrectomized rats, thus contributing to progression of CKD; Serum indole-3-acetic acid is a significant predictor of mortality and cardiovascular events in patients with CKD; induces pro-inflammatory enzyme COX-2 and oxidative stress | 71, 72 |
Phenylacetic acid (136 Da) | Endogenous; bacterial metabolism of tryptophan | Clostridum spp., Bacteroides spp. | Toxic; induces nausea, vomiting, diarrhea, and convulsion; associated with impaired immunoregulation, increased oxidative stress, and osteoblast dysfunction; shown to cause renal tubular damage in dogs | 88, 88–91 | |
Hippuric acid (179 Da) | Hippurate | Ingestion, bacterial metabolism of aromatic compounds and polyphenols | Clostridia spp. | Non-toxic; ↑ the anion gap acidosis; may cause glucose intolerance and interfere with erythropoisos and platelet cyclo-oxygenase activity | 94–96 |
COX-2 cyclooxygenase 2; CVD, cardiovascular disease; CKD, chronic kidney disease; MW, molecular weight; TMAO, Trimethylamine N Oxide
Ammonia and Urea
Inter-dependency between humans and microbes in the metabolic process is exemplified by the urea nitrogen salvage pathway. The end-product of mammalian protein catabolism is ammonia, which is toxic to cells in higher concentrations and thus converted to urea through the ornithine–urea cycle. Mammals cannot break down urea, but gut bacteria expressing urease cleave urea into ammonia and carbon dioxide.37 (Fig 3) Some of the ammonia can be utilized for microbial synthesis of amino acids or can enter the host circulation and serve as a substrate for synthetic processes.38 Ammonia in the large intestine could also be generated by microbial fermentation of glutamine, serine, threonine, and glycine. In a series of studies using CKD rats and human colonocytes, Vaziri et al. demonstrated that exposure to ammonia and ammonium hydroxide damages the intestinal epithelial tight junction and impairs its barrier function.39–41
Figure 3.
Schematic illustration of amino acid, ammonia (NH3), and urea flux between the gastrointestinal tract and liver. Urea is produced via the urea cycle in the liver from dietary amino acids and by their catabolism in peripheral tissues. Urea is then excreted into the gastrointestinal system and into the urine. Within the intestinal tract, gut bacteria, particularly coliforms and anaerobes in the colon and cecum, convert dietary amino acids and urea into ammonia and CO2, using microbial urease. Some of this ammonia is, in turn, converted to ammonium hydroxide which raises the luminal fluid’s pH before being excreted in feces. The remaining ammonia is absorbed into the portal circulation and converted back to urea via the urea cycle in the liver. Of the total ammonia produced, the majority enters the urea cycle, with the remaining smaller proportions being metabolized by peripheral tissues.
Creatinine, Guanidine and Uric Acid
Jones and Burnett estimated that about 16 to 60% of orally administered C14 labeled creatinine is either metabolized or excreted by routes other than the urine in subjects with kidney failure.42 Creatinine concentration in ileal effluent is similar to plasma, but it is undetectable in stool, suggesting that it is possibly degraded by colonic bacteria.43 Furthermore, 1-methyl guanidine is produced by the metabolism of creatinine by Pseudomonas stutzeri.43 Guanidine compounds accumulate in CKD, and some of them are considered uremic toxins.44 Administration of methyl guanidine to rats with kidney failure results in a dose-dependent increase in mortality.45
Urate (uric acid) is the end product of purine metabolism. Through excretion, the kidneys play an important role in maintaining serum urate levels, and sustained hyperuricemia in CKD is associated with gout, hypertension, and CVD.46, 47
Although the kidney is the primary route for the excretion of urate, there is a minimal rise in its serum concentration in advanced CKD due to CKD-induced adaptive secretion of uric acid by the colon.48 Consequently, bacterial families possessing urease, uricase, and enzymes capable of forming indole and p-cresol are expanded in ESRD patients.49
Indoles
Indoles are an aromatic group of compounds containing a pyrrole ring. Metabolism of tryptophan by bacterial tryptophanase generates more than 600 indoles in the gut, which are absorbed and sulfate conjugated in the liver.50
Indoxyl Sulfate (IS)
The concentration of IS in the serum is negatively correlated with the level of kidney function,51 and can be used as a predictor of CKD progression.52 IS is normally cleared by the proximal tubules of the kidneys, but accumulates in CKD patients. Cellular transport of IS is mediated by the organic anion transporter (OAT) 1 and OAT3,53 expression of which has been shown to be reduced in experimental models of kidney failure.54 Accumulation of IS in renal proximal tubular cells induces nephrotoxicity by activating nuclear factor (NF)-κB and plasminogen activator inhibitor type 1 expression.54, 55 Following its administration to uremic rats, IS increases the expression of genes such as tissue inhibitor of metalloproteinases and transforming growth factor (TGF)β1, which are known mediators of tubulointerstitial fibrosis.56
Elevated level of IS is associated with aortic calcification, vascular stiffness, and increased risk of overall and cardiovascular mortality in patients with CKD.57–59 Experimental studies show that IS enhances oxidative stress in endothelial cells,60 increases shedding of endothelial microparticles,61 impairs endothelial cell repair mechanism,62 and induces vascular smooth muscle cell proliferation.59 Interestingly, this indole may decrease erythropoietin production by interfering with oxygen sensing in erythropoietin producing cells.63 Furthermore, IS is taken up by osteoblasts, where it augments oxidative stress and down-regulates parathyroid hormone receptor expression, leading to low-turnover bone disease.64 IS also has an inhibitory effect on osteoclast function.65 In non-dialysis-dependent CKD patients, IS has been found to be positively associated with bone formation rate.66 IS is a ligand of the aryl-hydrocarbon receptor (AhR), a transcriptional regulator that has been shown to cause podocyte injury.67 Similar to p-cresol sulfate, IS is also highly protein bound, preventing it from being effectively removed by hemodialysis.68, 69
Indole Acetic Acid (IAA)
A protein-bound uremic solute, IAA is generated from tryptophan by both intestinal bacteria and normal cells. Plasma concentrations of IAA are elevated in CKD and the compound is only partly removed during hemodialysis.70 IAA has been shown to induce glomerular sclerosis and interstitial fibrosis in subtotally nephrectomized rats, thus contributing to progression of CKD.71 IAA is also a significant predictor of mortality and cardiovascular events in patients with CKD.72 IAA activates nongenomic AHR pathway, resulting in induction of the proinflammatory enzyme cyclooxygenase-2 and oxidative stress.72
Phenols
Phenols are aromatic compounds with one or more hydroxyl groups attached to a benzene ring. Partial breakdown of tyrosine and phenylalanine by several intestinal bacteria genera including Bacterioides, Bifidobacterium, Lactobacillus, Enterobacter, and Clostridium generate phenols and p-cresol.73 Most of the phenols produced in the colon are quickly absorbed and modified by sulfate, acetate, and—more rarely--glucuronide conjugation, especially in the liver or colonic mucosa, making them less toxic and facilitate their excretion by organic ion transport systems.74
p-cresol sulfate (PCS)
PCS is a 188-Da uremic retention solute and a uremic toxin.73, 75 Urinary excretion depends on tubular secretion through specific transporters,76 leading to progressive accumulation in patients with CKD.77 In experimental studies, IS and PCS activate the intrarenal renin-angiotensin system, TGF/Smad pathway, and possibly epithelial mesenchymal transformation leading to fibrosis of the kidney.78 Watanabe et al. showed that administration of PCS causes significant renal tubular damage in 5/6-nephrectomized rats by increasing oxidative stress and inflammatory cytokines.79 Elevated plasma PCS level is associated with all-cause mortality and CVD in patients with chronic kidney failure and earlier stages of CKD.80–82 Koppe et al.83 demonstrated the contribution of PCS to CKD-associated insulin resistance and cachexia by its action on adipose tissue and increase of lipolysis. Mice treated with PCS have been shown to display altered insulin signaling in skeletal muscle through activation of ERK1/2.
Phenylacetylglutamine (PAG)
PAG is a major microbe-derived nitrogenous metabolite that accumulates in uremia.84, 85 Most PGA is derived from β-phenylethylamine formed in the large intestine by decarboxylation of phenylalanine released by bacterial proteolysis of unabsorbed protein.86 PGA is also produced in the liver by metabolism of phenylacetic acid, derived from phenylalanine.87 The precursor phenylacetic acid itself is toxic and induces nausea, vomiting, diarrhea, and convulsion.88 The latter is associated with impaired immunoregulation,89 increased oxidative stress,90 and osteoblast dysfunction.91 Autopsy studies in dogs have shown that exposure to phenylacetic acid causes renal tubular damage, and thus may contribute to progression of kidney disease.88
Hippurate
Gut microbial metabolism generates benzoate from dietary aromatic compounds, and the subsequent hepatorenal conjugation of benzoate with glycine, forms hippurate.92, 93 Hippurate is generally believed to be non-toxic, except for contributing to the anion gap acidosis.94, 95 There is some evidence that hippurate may cause glucose intolerance and interfere with erythropoiesis and platelet cyclo-oxygenase activity.96
Amines
Polyamines
Generated by the gut microbiota from precursor amino acids, polyamines include putrescine, agmatine, cadaverine, tyramine, and histamine. Igarashi et al. observed a decrease in spermine and an increase in putrescine, as well as acrolein (a major toxic compound deriving from spermine), in the plasma of patients with CKD.97 Cellular downregulation by polyamines is proposed to play a role in the lack of tissue response to hormones in uremia.98 Polyamine also inhibit the activity of erythropoietin and may hence play a role in the anemia of CKD.99
D-amino Acids
Among other potential uremic toxins are some of the D-amino acids. It has been shown that plasma levels of certain D-amino acids increase as GFR declines.100 D-lactic acid originates from endogenous production by the methylglyoxylase pathway, or as a byproduct of bacterial metabolism in the intestine. Oh et al. first described D-lactic acidosis, which has been associated with neurotoxicity.101
RECENT ADVANCES
Trimethylamine N Oxide (TMAO)
In a landmark study, Wang et al. performed metabolomics studies and screened more than 2,000 compounds in 75 patients with CVD, and identified TMAO, choline, and betaine as being associated with heart disease.102 In a study involving 4,007 patients undergoing elective coronary angiography, elevated TMAO level was found to predict an increased risk of major adverse cardiovascular events after adjustment for traditional risk factors.8 Choline is catabolized by the intestinal microbiota to form trimethylamine gas, which is subsequently metabolized by the liver into TMAO. Dietary carnitine, which is found in red meat, is another substrate for gut flora to produce TMAO.103
Microbial taxa belonging to the Clostridiaceae and Peptostreptococcaceae families are positively associated with blood levels of TMAO in humans.103 The way in which TMAO promotes atherosclerosis remains speculative, but it has been shown to cause alteration of cholesterol and sterol metabolism, promote foam cell formation by increasing expression of scavenger receptors on macrophages, and lead to alterations in bile acid metabolism and sterol transporters in the liver and intestine.102, 103 High TMAO level is a predictor of increased long-term mortality in patients with heart failure, which is independent of traditional risk factors.104
Plasma level of TMAO is elevated in patients with CKD and is associated with increased risk of death.105 In animal models, feeding with TMAO and choline leads to tubulointerstitial fibrosis and collagen deposition, which is accompanied by a significant increase in the phosphorylation of Smad3.105
Hydrogen sulfide (H2S)
Sulfate-reducing bacteria in the human colon can use H2 or organic compounds as electron donors for reduction of sulfate or other oxidized sulfur compounds to generate H2S. This toxic gas belongs to the family of gasotransmitters and inhibits mitochondrial respiration through blockade of cytochrome c oxidase,106 with genotoxic, cytotoxic, and inflammatory effects.107 Despite these reports of toxicity, findings from animal models of ischemia/reperfusion injury and heart failure suggest that H2S could be cardioprotective.108 In animal models of CKD plasma, H2S production by the kidney and liver is reduced due to down-regulation of the H2S-producing enzymes.107 Preliminary evidence indicates that plasma H2S level is reduced in hemodialysis patients compared to controls.109 In in vitro studies, sodium hydrogen sulfide, a hydrogen sulfide donor, decreases inflammation and inhibited renal fibrosis, possibly through inhibiting the TGF-β1/Smad and mitogen-activated protein kinase signaling pathways.110 The potential beneficial effect of H2S in CKD and whether it retains its toxicity in higher concentration needs further study.
Endotoxin
Endotoxin is a phospholipid that forms the outer membrane of most Gram-negative bacteria. Circulating endotoxin binds lipopolysaccharide binding protein (LBP), forming a complex that interacts with the MD-2 part of the Toll-like receptor 4, anchored by CD14.111 This binding stimulates, via activation of NF-κB, the translation and production of inflammatory cytokines.112 Endotoxin translocation from the gut has been suggested as one of the causes of inflammation in CKD.103, 113 Furthermore, endotoxin is known to play an important role in initiation and progression of atherosclerosis by mediating endothelial cell injury, boosting recruitment of monocytes, transforming macrophages to foam cells, and activating coagulant activity.114, 115 We have demonstrated that soluble CD14 is associated with progression of CKD, CVD, and mortality in patients with kidney disease.116–118 Furthermore, endotoxin has been identified as an inflammatory trigger of insulin resistance, obesity, and diabetes in mice.119
Gut Microbe-Derived Mediators of Immune Regulation
It now evident that gut microbes play a key role in shaping of the human immune system.120 Polysaccharide A produced by Bacteroides fragilis induces accumulation of Foxp3-positive regulatory T cells and production of interleukin (IL)-10.121 Another molecule that is derived from gut microbiota and can modulate peripheral immune function is peptidoglycan, an essential component of the cell wall of virtually all bacteria. Upon entry to blood, peptidoglycan systemically primes the innate immune system.122 Peptidoglycan has been shown to signal via the pattern-recognition receptor Nod1.122 The role of these and other microbe-derived molecules in mediating dysregulated immune response in patients with CKD warrants further exploration.
Short-Chain Fatty Acids (SCFA)
An altered dysbiotic gut microbiome will not only produce an array of harmful metabolites and uremic toxins, but can also potentially cease to produce the otherwise beneficial metabolites such as SCFA. These one- to six-carbon aliphatic organic acids are the products of anaerobic bacterial fermentation of dietary polysaccharides, and include acetate, propionate, and butyrate. SCFA enter the systemic circulation through colonocytes by passive diffusion as well as active transport mechanisms. They affect a range of host functions, including energy metabolism, immune regulation and gut motility, and blood pressure regulation through activation of G protein-coupled receptors such as GPR41 and GPR43.123
Butyrate is the primary source of energy for colonocytes and is thus associated with maintenance of the epithelium. More recently, researchers have expanded the role of SCFA to explain the gut-kidney connection in ischemia reperfusion injury (IRI) showing that treatment with SCFA reduces kidney injury of this type in a germ-free mouse model system.124, 125 The key mechanism protecting against acute kidney injury was suggested to be reduction in inflammation mediated by an epigenetic mechanism. If SCFA are important in AKI, it might be of interest to also define their role in progression of CKD.
Interestingly, SCFA may also influence blood pressure through activation of GPR41 and GPR43, which are expressed in adipocytes, neutrophils, and sympathetic ganglia.126 Olfactory receptor 78 (Olfr78) expressed in the kidney responds to SCFA where it mediates renin secretion and increases blood pressure3 this increase is counteracted by GPR43, which induces vasodilatation.126
Advances in Technology and Discovery of Uremic Toxins
In 2003, the European Uremic Toxin Work Group catalogued 90 different uremic retention solutes,127 and this list has since grown in number to more than 150.128 The discovery of the newer solutes is primarily driven by the advent and implementation of “omic” technologies such as metagenomics, transcriptomics, proteomics, and metabolomics.129
It is apparent that uncultured microorganisms represent about 99% of the gut microbiota. Fortunately, since 16S ribosomal RNA sequences are highly conserved within organisms of the same genus and species, they can be used to determine phylogeny. Metagenomics is culture-independent analysis of microbial genomes, which allows understanding the dynamics and diversity of microbial community and its interaction with host.130
In the search for uremic toxins, use of proteomics and metabolomics could be complementary. Proteomics focuses on study of peptides and proteins, whereas metabolomics is useful in identification of small metabolites (<1000 Daltons) such as amino acids, alcohols, vitamins, polyols, organic acids, as well as nucleotides. Since metabolites are downstream of both transcription and translation, they may be more reflective of disturbed metabolism than proteins, messenger RNA, and genes, but each technique has a unique role in the discovery of uremic retention solutes.
Metabolite profiling of plasma samples from 1,434 Framingham study participants demonstrated that 9 metabolites predict the development of CKD.131 Interestingly, choline was one of the three markers that remained significant after adjustment for estimated glomerular filtration rate, age, sex, diabetes mellitus, hypertension, and proteinuria at baseline.131 A metabolomic study in stage 3–4 CKD patients revealed that 14 metabolites were elevated in uremic plasma.132 In addition to confirming the retention of several previously identified uremic toxins, including PCS, this study detected two novel uremic retentions solutes, dimethyl sulphone and 2-hydroxyisobutyric acid.132 It is essential to emphasize the importance of investigating the underlying biochemical mechanisms of any newly discovered uremic solute in order to determine its pathophysiologic importance.129
The Microbiome as a Therapeutic Target
Unlike the human genome, the gut microbiome has a dynamic composition that is susceptible to manipulation and selective “farming” of desired microbial populations for the benefit of the human host. As we unravel the details of interactions between the host and the microbiota, as well as those within the microbiota itself, new classes of therapeutics will emerge to harness the vast therapeutic potential of this extraordinary natural resource. Among the seemingly limitless potential applications of human gut microbiome, its use as disease biomarker, alteration of the microbiome composition to treat disease, and genetic engineering of the microbes to gain new functions or deliver small therapeutic molecules, have been proposed.
Gut microbiome-based biomarkers
Gut microbiome composition has been considered as a potential tool for diagnosing, monitoring and prognostication of diseases. Microbiome profile is not only altered in CKD, it also varies by underlying etiology.133–135 Whether the dysbiotic microbiome is a cause and/or consequence of CKD, is not quite clear.136 The potential utility of gut microbiome as a biomarker for screening individuals with susceptibility to develop CKD remains to be investigated.
Manipulation of the microbiome composition
An improved understanding of the gut microbiome’s physiologic functions and the pathologic effects of dysbiosis have triggered interest in various ways of reestablishing symbiosis.22 The generation of uremic toxins could be reduced by selectively increasing saccharolytic bacteria and reducing proteolytic bacteria in the colon. A number of therapeutic interventions have been explored to modulate gut microbiota, or adsorption of uremic toxin end products of microbial fermentation (Table 3).
Table 3.
Treatment options for uremic toxins and inflammation
Intervention | Patient type | Comments | Ref. |
---|---|---|---|
Probiotic | |||
Oligofructose-enriched inulin | Healthy participants (n=50) | ↓ Urinary excretion of p-cresol | 72 |
Inulin/oligofructose | Obese women (n=30) | ↓ Endotoxemia | 64 |
Oligosaccharides | Elderly participants (n=74) | ↓ TNF-α mRNA and IL-6 mRNA; Serum soluble CD14 | 73 |
Prebiotic | |||
Bifobacterium bifidum, B. catenulatum, B. longum, and Lactobacillus plantarum | Peritoneal dialysis patients (n=39; 21 in the probiotics group and 18 in the placebo group) | ↓ TNF-α, IL-5, IL-6, and endotoxin; ↑ IL-10; Preserve residual kidney function | 139 |
Oligofructose-enriched inulin | Hemodialysis patients (n=22) | ↓ Serum p-cresyl sulfate and generation rate | 58 |
Synbiotic | |||
Probiotic along with inulin, oat bran, pectin, and resistant starch | Trauma patients (n=65) | ↓ Rate of systemic inflammatory response, syndrome, infections, severe sepsis, and mortality | 74 |
Galacto-oligosaccharides and L. casei, and B. breve | Hemodialysis patients (n=7) | ↓ Serum p-cresol | 144, 167 |
Probinul neutro® | CKD patients stages 3–4 (n=30) | ↓ Total plasma p-cresol | 140 |
Oral adsorbent | |||
AST-120 | Adult patients with moderate to severe CKD (n=132)147; Adult patients with moderate to severe CKD (n=2035)149 | ↓ Indoxyl sulfate in a dose-dependent manner147; Unable to demonstrate a beneficial effect of AST-120 on progression of CKD149 | 147,149 |
α-galactosidase inhibitor | |||
Acarbose (an inhibitor of the α-glucosidase enzymes) | Healthy volunteers (n=9) | ↓Serum concentrations of p-cresol; ↓Urinary excretion of p-cresol; ↑Fecal excretion of nitrogen increased | 168 |
Genetically engineered bacteria | |||
Microencapsulated genetically engineered live E. coli DH5 cells | Uremic rats | ↓Plasma urea concentration; ↓Plasma ammonia concentration | 146 |
Ongoing studies | |||
SYNERGY - HMW inulin, fructooligosaccarides and galacto-oligosaccarides plus strains from the Lactobacillus, Bifidobacteria and Streptococcus genera | Patients with moderate to severe CKD stages 4–5 (n=37) | Ongoing study aiming to assess the effectiveness of synbiotics on the synthesis of uremic toxins indoxyl sulfate and p-cresol sulfate | 150 |
Arabinoxylan-oligosaccharides (AXOS) | Patients with CKD stage 3b-4 | Ongoing study aiming to assess whether AXOS can decrease intestinal generation and serum concentrations of microbial metabolites in patients with CKD | 169 |
CKD, chronic kidney disease; HMW, high-molecular-weight; IL, interleukin; mRNA, messenger RNA; TNF-α, tumor necrosis factor α;
Probiotics are defined as “live microorganisms” that when administered in adequate quantities bestow a health benefit on the host. However, administration of enteric capsule preparation of Bifidobacterium longum to patients with CKD was reported to have only minimal effect on CKD progression.137 In a randomized, double-blind, placebo-controlled crossover study involving 22 hemodialysis patients, Renadyl (a specific probiotic formulation) was not observed to have an effect on microbe-derived uremic toxins.138
Despite these negative findings and the absence of large-scale studies, a recent randomized, double-blind, placebo-controlled trial involving 39 peritoneal dialysis patients reported significant reduction in the serum levels of endotoxin and pro-inflammatory cytokines, increase in the serum levels of anti-inflammatory cytokine (IL-10), and preservation of residual kidney function following a 6 month probiotic therapy.139 In another double-blind, randomized placebo-controlled trial with 30 CKD patients (stages 3–4), the symbiotic Probinul neutro® was shown to significantly lower total plasma p-cresol concentrations after 2–4 weeks of treatment.140 One of the main limitations to probiotic therapy is that no study has yet demonstrated sustained survival of probiotics in the colon of dysbiotic CKD patients. Furthermore, care should be taken when choosing probiotics since the contribution of bacteria with the ability to hydrolyze urea may be more harmful.
Prebiotics are nondigestible food ingredients whose positive effects are due to stimulating the growth or activity of one or a limited number of bacteria in the colon. Meijers et al. showed that oligofructose inulin significantly reduces PCS generation rates and serum concentrations in hemodialysis patients, but has no effect on IS.141 In contrast, a randomized controlled trial showed that resistant starch decreased IS level in patients treated with hemodialysis.142 In rats, a high amylose-resistant starch diet was observed to retard CKD progression and attenuated oxidative stress and inflammation.143 A small study in hemodialysis patients showed that a synbiotic (combining Lactobacillus casei, Bifidobacterium breve and galacto-oligosaccharides) decreased p-cresol but not IS.144
Genetic engineering of the gut microbes
Genetic engineering of bacteria for delivery of drugs and small therapeutic molecules holds a tremendous potential as a future approach to treat a wide range of disease and promote health. Prakash and Chang used microencapsulated E. coli cells genetically engineered to contain a K. aerogenes gene, and showed that these modified bacteria were able to effectively remove urea and ammonia in vitro145 and lower plasma creatinine in rats when given orally on a daily basis.146 At present, safety concerns prevent the release of genetically engineered cells into the body, and the field anxiously welcomes further pioneering research to improve our knowledge on the gut microbial ecology so that it can make more educated attempts to manipulate the gut microbial community for therapeutic purposes.
Another method for decreasing gut-derived uremic toxins is to use oral sorbents. The oral sorbent AST-120 was found to decrease plasma IS in a dose-dependent manner.147 The primary effect of AST-120 appears to be mediated through its adsorption of urea-derived ammonia and interruption of enterohepatic urea recycling.148 Although small randomized controlled animal studies have suggested a renoprotective effect for AST-120, a subsequent large randomized controlled trial could not confirm it.149 The larger study has some methodological limitations, but it also raises the possibility that targeting specific microbiome-derived uremic toxin may not be sufficient, since the microbiomes generate a myriad of yet unidentified toxins.
Thus, the quality of evidence emerging from the small, uncontrolled mostly single-center studies targeting the microbiome needs careful scrutiny and these results need replication in well-designed large scale multicenter trials. Among the currently ongoing studies, some are worth mentioning. In a double-blind, placebo-controlled, randomized cross-over trial, Rossi et al., is recruiting 37 CKD patients for a 6 week-long synbiotic therapy (or placebo) to determine the efficacy of synbiotic therapy for lowering the serum IS and PCS.150 The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) is proposing to conduct two highly intensive phase II randomized, double-blind, placebo-controlled, multi-center studies examining the effect of prebiotic therapy on microbiome and the associated metabolomics profile in CKD patients not yet on dialysis and maintenance hemodialysis patients; these studies are expected to be launched shortly.
SUMMARY
It is becoming evident that interaction of the human host with our resident microbes at the gene, protein, and metabolite levels could have significant impact on health and disease. The features and composition of the gut flora reflects selective forces acting on both the microbial community and the host. Whether an adaptive change in microbiome in response to the uremic state becomes maladaptive, leading to increased generation of uremic toxins and associated complications, needs to be rigorously examined using modern techniques in clinical and experimental settings. It is also possible that the presence of specific microbiome or absence of protective microbiome increases the susceptibility to kidney disease when exposed to injury. (Fig 4) Supported by preliminary evidence and enticed by novelty of the concept, a number of interventions have been proposed to reduce uremic toxicity targeting the gut microbiome and tested with mixed results. The findings from these studies should be viewed cautiously and further examined in well-designed large scale studies prior to implementation. Needless to say, if proven to be effective, gut-microbiome targeted interventions will have a significant impact on the management of patients with chronic kidney failure and earlier stages of CKD.
Figure 4.
The cause-consequence relationship between gut dysbiosis and CKD. Human genome affects gut microbiome, and together, in presence of a specific dysbiotic microbiome or absence of protective microbiome, they can increases the susceptibility to kidney disease when exposed to insult. Subsequent maladaptive changes of microbiome in response to uremic state, and in association with traditional risk factors, lead to further increased generation of uremic toxins and disease progression.
Acknowledgments
Support: Dr Raj supported by National Institutes of Health grants 1R01DK073665-01A1, 1U01DK099924- 01, and 1U01DK099914-01.
Footnotes
Financial Disclosure: The authors declare that they have no relevant financial interests.
Peer Review: Evaluated by 2 external peer reviews, the Feature Editor, the Education Editor, and the Editor-in-Chief.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gevers D, Knight R, Petrosino JF, et al. The Human Microbiome Project: a community resource for the healthy human microbiome. PLoS Biol. 2012;10(8):e1001377. doi: 10.1371/journal.pbio.1001377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915–1920. doi: 10.1126/science.1104816. [DOI] [PubMed] [Google Scholar]
- 4.Lederberg J. Infectious history. Science. 2000;288(5464):287–293. doi: 10.1126/science.288.5464.287. [DOI] [PubMed] [Google Scholar]
- 5.Hawrelak JA, Myers SP. The causes of intestinal dysbiosis: a review. Altern Med Rev. 2004;9(2):180–197. [PubMed] [Google Scholar]
- 6.Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101(44):15718–15723. doi: 10.1073/pnas.0407076101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
- 8.Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–1584. doi: 10.1056/NEJMoa1109400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr. 2002;22:283–307. doi: 10.1146/annurev.nutr.22.011602.092259. [DOI] [PubMed] [Google Scholar]
- 10.Hill MJ. Intestinal flora and endogenous vitamin synthesis. Eur J Cancer Prev. 1997;6(Suppl 1):S43–S45. doi: 10.1097/00008469-199703001-00009. [DOI] [PubMed] [Google Scholar]
- 11.Hylemon PB, Harder J. Biotransformation of monoterpenes, bile acids, and other isoprenoids in anaerobic ecosystems. FEMS Microbiol Rev. 1998;22(5):475–488. doi: 10.1111/j.1574-6976.1998.tb00382.x. [DOI] [PubMed] [Google Scholar]
- 12.Duncan SH, Richardson AJ, Kaul P, Holmes RP, Allison MJ, Stewart CS. Oxalobacter formigenes and its potential role in human health. Appl Environ Microbiol. 2002;68(8):3841–3847. doi: 10.1128/AEM.68.8.3841-3847.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Braun-Fahrlander C, Riedler J, Herz U, et al. Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med. 2002;347(12):869–877. doi: 10.1056/NEJMoa020057. [DOI] [PubMed] [Google Scholar]
- 14.Xu J, Gordon JI. Honor thy symbionts. Proc Natl Acad Sci U S A. 2003;100(18):10452–10459. doi: 10.1073/pnas.1734063100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.A framework for human microbiome research. Nature. 2012;486(7402):215–221. doi: 10.1038/nature11209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Structure function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–214. doi: 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308(5728):1635–1638. doi: 10.1126/science.1110591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489(7415):242–249. doi: 10.1038/nature11552. [DOI] [PubMed] [Google Scholar]
- 19.Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi: 10.1038/nature11053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Muegge BD, Kuczynski J, Knights D, et al. Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science. 2011;332(6032):970–974. doi: 10.1126/science.1198719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Metchnikoff E. Essais optimistes. Paris. In: Chalmers Mitchell P, translator and editor. The Prolongation of Life: Optimistic Studies. London: Heinemann; 1907. [Google Scholar]
- 22.Ramezani A, Raj DS. The gut microbiome, kidney disease, and targeted interventions. J Am Soc Nephrol. 2014;25(4):657–670. doi: 10.1681/ASN.2013080905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Vaziri ND, Wong J, Pahl M, et al. Chronic kidney disease alters intestinal microbial flora. Kidney Int. 2013;83(2):308–315. doi: 10.1038/ki.2012.345. [DOI] [PubMed] [Google Scholar]
- 24.Hida M, Aiba Y, Sawamura S, Suzuki N, Satoh T, Koga Y. Inhibition of the accumulation of uremic toxins in the blood and their precursors in the feces after oral administration of Lebenin, a lactic acid bacteria preparation, to uremic patients undergoing hemodialysis. Nephron. 1996;74(2):349–355. doi: 10.1159/000189334. [DOI] [PubMed] [Google Scholar]
- 25.Wu MJ, Chang CS, Cheng CH, et al. Colonic transit time in long-term dialysis patients. Am J Kidney Dis. 2004;44(2):322–327. doi: 10.1053/j.ajkd.2004.04.048. [DOI] [PubMed] [Google Scholar]
- 26.Bammens B, Verbeke K, Vanrenterghem Y, Evenepoel P. Evidence for impaired assimilation of protein in chronic renal failure. Kidney Int. 2003;64(6):2196–2203. doi: 10.1046/j.1523-1755.2003.00314.x. [DOI] [PubMed] [Google Scholar]
- 27.Krishnamurthy VM, Wei G, Baird BC, et al. High dietary fiber intake is associated with decreased inflammation and all-cause mortality in patients with chronic kidney disease. Kidney Int. 2012;81(3):300–306. doi: 10.1038/ki.2011.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wandersman C, Delepelaire P. Bacterial iron sources: from siderophores to hemophores. Annu Rev Microbiol. 2004;58:611–647. doi: 10.1146/annurev.micro.58.030603.123811. [DOI] [PubMed] [Google Scholar]
- 29.Jernberg C, Lofmark S, Edlund C, Jansson JK. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology. 2010;156(Pt 11):3216–3223. doi: 10.1099/mic.0.040618-0. [DOI] [PubMed] [Google Scholar]
- 30.Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4554–4561. doi: 10.1073/pnas.1000087107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6(11):e280. doi: 10.1371/journal.pbio.0060280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Einheber A, Carter D. The role of the microbial flora in uremia. I. Survival times of germfree, limited-flora, and conventionalized rats after bilateral nephrectomy and fasting. J Exp Med. 1966;123(2):239–250. doi: 10.1084/jem.123.2.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Aronov PA, Luo FJ, Plummer NS, et al. Colonic contribution to uremic solutes. J Am Soc Nephrol. 2011;22(9):1769–1776. doi: 10.1681/ASN.2010121220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wikoff WR, Anfora AT, Liu J, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106(10):3698–3703. doi: 10.1073/pnas.0812874106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sorensen LB. Role of the intestinal tract in the elimination of uric acid. Arthritis Rheum. 1965;8(5):694–706. doi: 10.1002/art.1780080429. [DOI] [PubMed] [Google Scholar]
- 36.Gibson SA, McFarlan C, Hay S, Macfarlane GT. Significance of microflora in proteolysis in the colon. Appl Environ Microbiol. 1989;55(3):679–683. doi: 10.1128/aem.55.3.679-683.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Walser M, BODENLOS LJ. Urea metabolism in man. J Clin Invest. 1959;38:1617–1626. doi: 10.1172/JCI103940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Stewart GS, Smith CP. Urea nitrogen salvage mechanisms and their relevance to ruminants, non-ruminants and man. Nutr Res Rev. 2005;18(1):49–62. doi: 10.1079/NRR200498. [DOI] [PubMed] [Google Scholar]
- 39.Vaziri ND, Yuan J, Rahimi A, Ni Z, Said H, Subramanian VS. Disintegration of colonic epithelial tight junction in uremia: a likely cause of CKD-associated inflammation. Nephrol Dial Transplant. 2012;27(7):2686–2693. doi: 10.1093/ndt/gfr624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Vaziri ND, Goshtasbi N, Yuan J, et al. Uremic plasma impairs barrier function and depletes the tight junction protein constituents of intestinal epithelium. Am J Nephrol. 2012;36(5):438–443. doi: 10.1159/000343886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Vaziri ND, Yuan J, Norris K. Role of urea in intestinal barrier dysfunction and disruption of epithelial tight junction in chronic kidney disease. Am J Nephrol. 2013;37(1):1–6. doi: 10.1159/000345969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jones JD, Burnett PC. Creatinine metabolism in humans with decreased renal function: creatinine deficit. Clin Chem. 1974;20(9):1204–1212. [PubMed] [Google Scholar]
- 43.Van Eyk HG, Vermaat RJ, Leijnse-Ybema HJ, Leijnse B. The conversion of creatinine by creatiniase of bacterial origin. Enzymologia. 1986;34:198–202. [PubMed] [Google Scholar]
- 44.OLSEN NS, BASSETT JW. Blood levels of urea nitrogen, phenol, guanidine and creatinine in uremia. Am J Med. 1951;10(1):52–59. doi: 10.1016/0002-9343(51)90218-5. [DOI] [PubMed] [Google Scholar]
- 45.Yokozawa T, Mo ZL, Oura H. Comparison of toxic effects of methylguanidine, guanidinosuccinic acid and creatinine in rats with adenine-induced chronic renal failure. Nephron. 1989;51(3):388–392. doi: 10.1159/000185328. [DOI] [PubMed] [Google Scholar]
- 46.Alderman MH, Cohen H, Madhavan S, Kivlighn S. Serum uric acid and cardiovascular events in successfully treated hypertensive patients. Hypertension. 1999;34(1):144–150. doi: 10.1161/01.hyp.34.1.144. [DOI] [PubMed] [Google Scholar]
- 47.Gibson T, Highton J, Potter C, Simmonds HA. Renal impairment and gout. Ann Rheum Dis. 1980;39(5):417–423. doi: 10.1136/ard.39.5.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hatch M, Vaziri ND. Enhanced enteric excretion of urate in rats with chronic renal failure. Clin Sci (Lond) 1994;86(5):511–516. doi: 10.1042/cs0860511. [DOI] [PubMed] [Google Scholar]
- 49.Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol. 2014;39(3):230–237. doi: 10.1159/000360010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.van Haard PM. Chromatography of urinary indole derivatives. J Chromatogr. 1988;429:59–94. doi: 10.1016/s0378-4347(00)83867-0. [DOI] [PubMed] [Google Scholar]
- 51.Lin CJ, Chen HH, Pan CF, et al. p-Cresylsulfate and indoxyl sulfate level at different stages of chronic kidney disease. J Clin Lab Anal. 2011;25(3):191–197. doi: 10.1002/jcla.20456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wu IW, Hsu KH, Lee CC, et al. p-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol Dial Transplant. 2011;26(3):938–947. doi: 10.1093/ndt/gfq580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Motojima M, Hosokawa A, Yamato H, Muraki T, Yoshioka T. Uraemic toxins induce proximal tubular injury via organic anion transporter 1-mediated uptake. Br J Pharmacol. 2002;135(2):555–563. doi: 10.1038/sj.bjp.0704482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Motojima M, Hosokawa A, Yamato H, Muraki T, Yoshioka T. Uremic toxins of organic anions up-regulate PAI-1 expression by induction of NF-kappaB and free radical in proximal tubular cells. Kidney Int. 2003;63(5):1671–1680. doi: 10.1046/j.1523-1755.2003.00906.x. [DOI] [PubMed] [Google Scholar]
- 55.Lysaght MJ, Vonesh EF, Gotch F, et al. The influence of dialysis treatment modality on the decline of remaining renal function. ASAIO Trans. 1991;37(4):598–604. [PubMed] [Google Scholar]
- 56.Miyazaki T, Ise M, Seo H, Niwa T. Indoxyl sulfate increases the gene expressions of TGF-beta 1, TIMP-1 and pro-alpha 1(I) collagen in uremic rat kidneys. Kidney Int Suppl. 1997;62:S15–S22. [PubMed] [Google Scholar]
- 57.Adijiang A, Higuchi Y, Nishijima F, Shimizu H, Niwa T. Indoxyl sulfate, a uremic toxin, promotes cell senescence in aorta of hypertensive rats. Biochem Biophys Res Commun. 2010;399(4):637–641. doi: 10.1016/j.bbrc.2010.07.130. [DOI] [PubMed] [Google Scholar]
- 58.Barreto FC, Barreto DV, Liabeuf S, et al. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol. 2009;4(10):1551–1558. doi: 10.2215/CJN.03980609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yamamoto H, Tsuruoka S, Ioka T, et al. Indoxyl sulfate stimulates proliferation of rat vascular smooth muscle cells. Kidney Int. 2006;69(10):1780–1785. doi: 10.1038/sj.ki.5000340. [DOI] [PubMed] [Google Scholar]
- 60.Dou L, Jourde-Chiche N, Faure V, et al. The uremic solute indoxyl sulfate induces oxidative stress in endothelial cells. J Thromb Haemost. 2007;5(6):1302–1308. doi: 10.1111/j.1538-7836.2007.02540.x. [DOI] [PubMed] [Google Scholar]
- 61.Amabile N, Guerin AP, Leroyer A, et al. Circulating endothelial microparticles are associated with vascular dysfunction in patients with end-stage renal failure. J Am Soc Nephrol. 2005;16(11):3381–3388. doi: 10.1681/ASN.2005050535. [DOI] [PubMed] [Google Scholar]
- 62.Dou L, Bertrand E, Cerini C, et al. The uremic solutes p-cresol and indoxyl sulfate inhibit endothelial proliferation and wound repair. Kidney Int. 2004;65(2):442–451. doi: 10.1111/j.1523-1755.2004.00399.x. [DOI] [PubMed] [Google Scholar]
- 63.Chiang CK, Tanaka T, Inagi R, Fujita T, Nangaku M. Indoxyl sulfate, a representative uremic toxin, suppresses erythropoietin production in a HIF-dependent manner. Lab Invest. 2011;91(11):1564–1571. doi: 10.1038/labinvest.2011.114. [DOI] [PubMed] [Google Scholar]
- 64.Nii-Kono T, Iwasaki Y, Uchida M, et al. Indoxyl sulfate induces skeletal resistance to parathyroid hormone in cultured osteoblastic cells. Kidney Int. 2007;71(8):738–743. doi: 10.1038/sj.ki.5002097. [DOI] [PubMed] [Google Scholar]
- 65.Mozar A, Louvet L, Godin C, et al. Indoxyl sulphate inhibits osteoclast differentiation and function. Nephrol Dial Transplant. 2012;27(6):2176–2181. doi: 10.1093/ndt/gfr647. [DOI] [PubMed] [Google Scholar]
- 66.Barreto FC, Barreto DV, Canziani ME, et al. Association between indoxyl sulfate and bone histomorphometry in pre-dialysis chronic kidney disease patients. J Bras Nefrol. 2014;36(3):289–296. doi: 10.5935/0101-2800.20140042. [DOI] [PubMed] [Google Scholar]
- 67.Ichii O, Otsuka-Kanazawa S, Nakamura T, et al. Podocyte injury caused by indoxyl sulfate, a uremic toxin and aryl-hydrocarbon receptor ligand. PLoS One. 2014;9(9):e108448. doi: 10.1371/journal.pone.0108448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Niwa T, Takeda N, Tatematsu A, Maeda K. Accumulation of indoxyl sulfate, an inhibitor of drug-binding, in uremic serum as demonstrated by internal-surface reversed-phase liquid chromatography. Clin Chem. 1988;34(11):2264–2267. [PubMed] [Google Scholar]
- 69.Martinez AW, Recht NS, Hostetter TH, Meyer TW. Removal of P-cresol sulfate by hemodialysis. J Am Soc Nephrol. 2005;16(11):3430–3436. doi: 10.1681/ASN.2005030310. [DOI] [PubMed] [Google Scholar]
- 70.De SR, Dhondt A, Eloot S, Galli F, Waterloos MA, Vanholder R. Effect of the super-flux cellulose triacetate dialyser membrane on the removal of non-protein-bound and protein-bound uraemic solutes. Nephrol Dial Transplant. 2007;22(7):2006–2012. doi: 10.1093/ndt/gfm065. [DOI] [PubMed] [Google Scholar]
- 71.Satoh M, Hayashi H, Watanabe M, et al. Uremic toxins overload accelerates renal damage in a rat model of chronic renal failure. Nephron Exp Nephrol. 2003;95(3):e111–e118. doi: 10.1159/000074327. [DOI] [PubMed] [Google Scholar]
- 72.Dou L, Sallee M, Cerini C, et al. The cardiovascular effect of the uremic solute indole-3 acetic acid. J Am Soc Nephrol. 2015;26(4):876–887. doi: 10.1681/ASN.2013121283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Cummings JH. Fermentation in the human large intestine: evidence and implications for health. Lancet. 1983;1(8335):1206–1209. doi: 10.1016/s0140-6736(83)92478-9. [DOI] [PubMed] [Google Scholar]
- 74.De LH, Bammens B, Evenepoel P, De PV, Verbeke K. Gas chromatographic-mass spectrometric analysis for measurement of p-cresol and its conjugated metabolites in uremic and normal serum. Clin Chem. 2005;51(8):1535–1538. doi: 10.1373/clinchem.2005.050781. [DOI] [PubMed] [Google Scholar]
- 75.Meijers BK, Evenepoel P. The gut-kidney axis: indoxyl sulfate, p-cresyl sulfate and CKD progression. Nephrol Dial Transplant. 2011;26(3):759–761. doi: 10.1093/ndt/gfq818. [DOI] [PubMed] [Google Scholar]
- 76.Mutsaers HA, Wilmer MJ, van den Heuvel LP, Hoenderop JG, Masereeuw R. Basolateral transport of the uraemic toxin p-cresyl sulfate: role for organic anion transporters? Nephrol Dial Transplant. 2011;26(12):4149. doi: 10.1093/ndt/gfr562. [DOI] [PubMed] [Google Scholar]
- 77.Poesen R, Viaene L, Verbeke K, et al. Renal clearance and intestinal generation of p-cresyl sulfate and indoxyl sulfate in CKD. Clin J Am Soc Nephrol. 2013;8(9):1508–1514. doi: 10.2215/CJN.00300113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sun CY, Chang SC, Wu MS. Uremic toxins induce kidney fibrosis by activating intrarenal renin-angiotensin-aldosterone system associated epithelial-to-mesenchymal transition. PLoS One. 2012;7(3):e34026. doi: 10.1371/journal.pone.0034026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Watanabe H, Miyamoto Y, Honda D, et al. p-Cresyl sulfate causes renal tubular cell damage by inducing oxidative stress by activation of NADPH oxidase. Kidney Int. 2013;83(4):582–592. doi: 10.1038/ki.2012.448. [DOI] [PubMed] [Google Scholar]
- 80.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(6):1081–1087. doi: 10.1038/sj.ki.5000115. [DOI] [PubMed] [Google Scholar]
- 81.Liabeuf S, Barreto DV, Barreto FC, et al. Free p-cresylsulphate is a predictor of mortality in patients at different stages of chronic kidney disease. Nephrol Dial Transplant. 2010;25(4):1183–1191. doi: 10.1093/ndt/gfp592. [DOI] [PubMed] [Google Scholar]
- 82.Meijers BK, Claes K, Bammens B, et al. p-Cresol and cardiovascular risk in mild-to-moderate kidney disease. Clin J Am Soc Nephrol. 2010;5(7):1182–1189. doi: 10.2215/CJN.07971109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Koppe L, Pillon NJ, Vella RE, et al. p-Cresyl sulfate promotes insulin resistance associated with CKD. J Am Soc Nephrol. 2013;24(1):88–99. doi: 10.1681/ASN.2012050503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zimmerman L, Egestad B, Jornvall H, Bergstrom J. Identification and determination of phenylacetylglutamine, a major nitrogenous metabolite in plasma of uremic patients. Clin Nephrol. 1989;32(3):124–128. [PubMed] [Google Scholar]
- 85.Smith EA, Macfarlane GT. Formation of Phenolic and Indolic Compounds by Anaerobic Bacteria in the Human Large Intestine. Microb Ecol. 1997;33(3):180–188. doi: 10.1007/s002489900020. [DOI] [PubMed] [Google Scholar]
- 86.Seakins JW. The determination of urinary phenylacetylglutamine as phenylacetic acid. Studies on its origin in normal subjects and children with cystic fibrosis. Clin Chim Acta. 1971;35(1):121–131. doi: 10.1016/0009-8981(71)90302-0. [DOI] [PubMed] [Google Scholar]
- 87.Yang D, Beylot M, Agarwal KC, Soloviev MV, Brunengraber H. Assay of the human liver citric acid cycle probe phenylacetylglutamine and of phenylacetate in plasma by gas chromatography-mass spectrometry. Anal Biochem. 1993;212(1):277–282. doi: 10.1006/abio.1993.1323. [DOI] [PubMed] [Google Scholar]
- 88.Sherwin CP, Kennard S. Toxicity of phenylacetic acid. J Biol Chem. 1919;12:259–264. [Google Scholar]
- 89.Schmidt S, Westhoff TH, Krauser P, et al. The uraemic toxin phenylacetic acid impairs macrophage function. Nephrol Dial Transplant. 2008;23(11):3485–3493. doi: 10.1093/ndt/gfn266. [DOI] [PubMed] [Google Scholar]
- 90.Schmidt S, Westhoff TH, Krauser P, Zidek W, van der Giet M. The uraemic toxin phenylacetic acid increases the formation of reactive oxygen species in vascular smooth muscle cells. Nephrol Dial Transplant. 2008;23(1):65–71. doi: 10.1093/ndt/gfm475. [DOI] [PubMed] [Google Scholar]
- 91.Yano S, Yamaguchi T, Kanazawa I, et al. The uraemic toxin phenylacetic acid inhibits osteoblastic proliferation and differentiation: an implication for the pathogenesis of low turnover bone in chronic renal failure. Nephrol Dial Transplant. 2007;22(11):3160–3165. doi: 10.1093/ndt/gfm455. [DOI] [PubMed] [Google Scholar]
- 92.Remer T, Manz F. Paleolithic diet, sweet potato eaters, and potential renal acid load. Am J Clin Nutr. 2003;78(4):802–803. doi: 10.1093/ajcn/78.4.802. [DOI] [PubMed] [Google Scholar]
- 93.Li M, Wang B, Zhang M, et al. Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci U S A. 2008;105(6):2117–2122. doi: 10.1073/pnas.0712038105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Cathcart-Rake W, Porter R, Whittier F, Stein P, Carey M, Grantham J. Effect of diet on serum accumulation and renal excretion of aryl acids and secretory activity in normal and uremic man. Am J Clin Nutr. 1975;28(10):1110–1115. doi: 10.1093/ajcn/28.10.1110. [DOI] [PubMed] [Google Scholar]
- 95.Mitch WE, Brusilow S. Benzoate-induced changes in glycine and urea metabolism in patients with chronic renal failure. J Pharmacol Exp Ther. 1982;222(3):572–575. [PubMed] [Google Scholar]
- 96.Ringoir S, Vanholder R, Massy Z. Proceedings of the Ghent Symposium on Uremic Toxins. Advances in Experimental Medicine and Biology. 1986;223:59–67. [Google Scholar]
- 97.Igarashi K, Ueda S, Yoshida K, Kashiwagi K. Polyamines in renal failure. Amino Acids. 2006;31(4):477–483. doi: 10.1007/s00726-006-0264-7. [DOI] [PubMed] [Google Scholar]
- 98.Campbell RA, Grettie DP, Bartos F, Bartos D, Marton LJ. Uremic polyamine dysmetabolism. Proc Clin Dial Transplant Forum. 1978;8:194–198. [PubMed] [Google Scholar]
- 99.Kushner D, Beckman B, Nguyen L, et al. Polyamines in the anemia of end-stage renal disease. Kidney Int. 1991;39(4):725–732. doi: 10.1038/ki.1991.88. [DOI] [PubMed] [Google Scholar]
- 100.Bruckner H, Hausch M. Gas chromatographic characterization of free D-amino acids in the blood serum of patients with renal disorders and of healthy volunteers. J Chromatogr. 1993;614(1):7–17. doi: 10.1016/0378-4347(93)80218-s. [DOI] [PubMed] [Google Scholar]
- 101.Oh MS, Phelps KR, Traube M, Barbosa-Saldivar JL, Boxhill C, Carroll HJ. D-lactic acidosis in a man with the short-bowel syndrome. N Engl J Med. 1979;301(5):249–252. doi: 10.1056/NEJM197908023010505. [DOI] [PubMed] [Google Scholar]
- 102.Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63. doi: 10.1038/nature09922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–585. doi: 10.1038/nm.3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tang WH, Wang Z, Fan Y, et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol. 2014;64(18):1908–1914. doi: 10.1016/j.jacc.2014.02.617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tang WH, Wang Z, Kennedy DJ, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res. 2015;116(3):448–455. doi: 10.1161/CIRCRESAHA.116.305360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Reiffenstein RJ, Hulbert WC, Roth SH. Toxicology of hydrogen sulfide. Annu Rev Pharmacol Toxicol. 1992;32:109–134. doi: 10.1146/annurev.pa.32.040192.000545. [DOI] [PubMed] [Google Scholar]
- 107.Aminzadeh MA, Vaziri ND. Downregulation of the renal and hepatic hydrogen sulfide (H2S)-producing enzymes and capacity in chronic kidney disease. Nephrol Dial Transplant. 2012;27(2):498–504. doi: 10.1093/ndt/gfr560. [DOI] [PubMed] [Google Scholar]
- 108.Perna AF, Lanza D, Sepe I, et al. Hydrogen sulfide, a toxic gas with cardiovascular properties in uremia: how harmful is it? Blood Purif. 2011;31(1–3):102–106. doi: 10.1159/000321838. [DOI] [PubMed] [Google Scholar]
- 109.Perna AF, Luciano MG, Ingrosso D, et al. Hydrogen sulphide-generating pathways in haemodialysis patients: a study on relevant metabolites and transcriptional regulation of genes encoding for key enzymes. Nephrol Dial Transplant. 2009;24(12):3756–3763. doi: 10.1093/ndt/gfp378. [DOI] [PubMed] [Google Scholar]
- 110.Song K, Wang F, Li Q, et al. Hydrogen sulfide inhibits the renal fibrosis of obstructive nephropathy. Kidney Int. 2014;85(6):1318–1329. doi: 10.1038/ki.2013.449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Pugin J, Heumann ID, Tomasz A, et al. CD14 is a pattern recognition receptor. Immunity. 1994;1(6):509–516. doi: 10.1016/1074-7613(94)90093-0. [DOI] [PubMed] [Google Scholar]
- 112.Freudenberg MA, Tchaptchet S, Keck S, et al. Lipopolysaccharide sensing an important factor in the innate immune response to Gram-negative bacterial infections: benefits and hazards of LPS hypersensitivity. Immunobiology. 2008;213(3–4):193–203. doi: 10.1016/j.imbio.2007.11.008. [DOI] [PubMed] [Google Scholar]
- 113.Freudenberg MA, Tchaptchet S, Keck S, et al. Lipopolysaccharide sensing an important factor in the innate immune response to Gram-negative bacterial infections: benefits and hazards of LPS hypersensitivity. Immunobiology. 2008;213(3–4):193–203. doi: 10.1016/j.imbio.2007.11.008. [DOI] [PubMed] [Google Scholar]
- 114.Eggesbo JB, Hjermann I, Ovstebo R, Joo GB, Kierulf P. LPS induced procoagulant activity and plasminogen activator activity in mononuclear cells from persons with high or low levels of HDL lipoprotein. Thromb Res. 1995;77(5):441–452. doi: 10.1016/0049-3848(95)93880-9. [DOI] [PubMed] [Google Scholar]
- 115.Reidy MA, Bowyer DE. Distortion of endothelial repair. The effect of hypercholesterolaemia on regeneration of aortic endothelium following injury by endotoxin. A scanning electron microscope study. Atherosclerosis. 1978;29(4):459–466. doi: 10.1016/0021-9150(78)90174-0. [DOI] [PubMed] [Google Scholar]
- 116.Poesen R, Ramezani A, Claes K, et al. Associations of Soluble CD14 and Endotoxin with Mortality, Cardiovascular Disease, and Progression of Kidney Disease among Patients with CKD. Clin J Am Soc Nephrol. 2015 doi: 10.2215/CJN.03100315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Raj DS, Carrero JJ, Shah VO, et al. Soluble CD14 levels, interleukin 6, and mortality among prevalent hemodialysis patients. Am J Kidney Dis. 2009;54(6):1072–1080. doi: 10.1053/j.ajkd.2009.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Raj DS, Shah VO, Rambod M, Kovesdy CP, Kalantar-Zadeh K. Association of soluble endotoxin receptor CD14 and mortality among patients undergoing hemodialysis. Am J Kidney Dis. 2009;54(6):1062–1071. doi: 10.1053/j.ajkd.2009.06.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761–1772. doi: 10.2337/db06-1491. [DOI] [PubMed] [Google Scholar]
- 120.Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol. 2004;4(6):478–485. doi: 10.1038/nri1373. [DOI] [PubMed] [Google Scholar]
- 121.Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A. 2010;107(27):12204–12209. doi: 10.1073/pnas.0909122107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat Med. 2010;16(2):228–231. doi: 10.1038/nm.2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Layden BT, Angueira AR, Brodsky M, Durai V, Lowe WL., Jr Short chain fatty acids and their receptors: new metabolic targets. Transl Res. 2013;161(3):131–140. doi: 10.1016/j.trsl.2012.10.007. [DOI] [PubMed] [Google Scholar]
- 124.Andrade-Oliveira V, Amano MT, Correa-Costa M, et al. Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. J Am Soc Nephrol. 2015;26(8):1877–1888. doi: 10.1681/ASN.2014030288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Barrows IR, Ramezani A, Raj DS. Gut Feeling in AKI: The Long Arm of Short-Chain Fatty Acids. J Am Soc Nephrol. 2015;26(8):1755–1757. doi: 10.1681/ASN.2014111157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Pluznick JL, Protzko RJ, Gevorgyan H, et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci U S A. 2013;110(11):4410–4415. doi: 10.1073/pnas.1215927110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Vanholder R, Argiles A, Baurmeister U, et al. Uremic toxicity: present state of the art. Int J Artif Organs. 2001;24(10):695–725. [PubMed] [Google Scholar]
- 128.Duranton F, Cohen G, De SR, et al. Normal and pathologic concentrations of uremic toxins. J Am Soc Nephrol. 2012;23(7):1258–1270. doi: 10.1681/ASN.2011121175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Vanholder R, Boelaert J, Glorieux G, Eloot S. New methods and technologies for measuring uremic toxins and quantifying dialysis adequacy. Semin Dial. 2015;28(2):114–124. doi: 10.1111/sdi.12331. [DOI] [PubMed] [Google Scholar]
- 130.Tringe SG, Rubin EM. Metagenomics: DNA sequencing of environmental samples. Nat Rev Genet. 2005;6(11):805–814. doi: 10.1038/nrg1709. [DOI] [PubMed] [Google Scholar]
- 131.Rhee EP, Clish CB, Ghorbani A, et al. A combined epidemiologic and metabolomic approach improves CKD prediction. J Am Soc Nephrol. 2013;24(8):1330–1338. doi: 10.1681/ASN.2012101006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Mutsaers HA, Engelke UF, Wilmer MJ, et al. Optimized metabolomic approach to identify uremic solutes in plasma of stage 3–4 chronic kidney disease patients. PLoS One. 2013;8(8):e71199. doi: 10.1371/journal.pone.0071199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60. doi: 10.1038/nature11450. [DOI] [PubMed] [Google Scholar]
- 134.Hevia A, Milani C, Lopez P, et al. Intestinal dysbiosis associated with systemic lupus erythematosus. MBio. 2014;5(5):e01548–14. doi: 10.1128/mBio.01548-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.De AM, Montemurno E, Piccolo M, et al. Microbiota and metabolome associated with immunoglobulin A nephropathy (IgAN) PLoS One. 2014;9(6):e99006. doi: 10.1371/journal.pone.0099006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Miyamoto Y, Watanabe H, Noguchi T, et al. Organic anion transporters play an important role in the uptake of p-cresyl sulfate, a uremic toxin, in the kidney. Nephrol Dial Transplant. 2011;26(8):2498–2502. doi: 10.1093/ndt/gfq785. [DOI] [PubMed] [Google Scholar]
- 137.Ando Y, Miyata Y, Tanba K, et al. Effect of oral intake of an enteric capsule preparation containing Bifidobacterium longum on the progression of chronic renal failure. Nihon Jinzo Gakkai Shi. 2003;45(8):759–764. [PubMed] [Google Scholar]
- 138.Natarajan R, Pechenyak B, Vyas U, et al. Randomized controlled trial of strain-specific probiotic formulation (Renadyl) in dialysis patients. Biomed Res Int. 2014;2014:568571. doi: 10.1155/2014/568571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Wang IK, Wu YY, Yang YF, et al. The effect of probiotics on serum levels of cytokine and endotoxin in peritoneal dialysis patients: a randomised, double-blind, placebo-controlled trial. Benef Microbes. 2015;6(4):423–430. doi: 10.3920/BM2014.0088. [DOI] [PubMed] [Google Scholar]
- 140.Guida B, Germano R, Trio R, et al. Effect of short-term synbiotic treatment on plasma p-cresol levels in patients with chronic renal failure: a randomized clinical trial. Nutr Metab Cardiovasc Dis. 2014;24(9):1043–1049. doi: 10.1016/j.numecd.2014.04.007. [DOI] [PubMed] [Google Scholar]
- 141.Meijers BK, De PV, Verbeke K, Vanrenterghem Y, Evenepoel P. p-Cresyl sulfate serum concentrations in haemodialysis patients are reduced by the prebiotic oligofructose-enriched inulin. Nephrol Dial Transplant. 2010;25(1):219–224. doi: 10.1093/ndt/gfp414. [DOI] [PubMed] [Google Scholar]
- 142.Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW. Effect of increasing dietary fiber on plasma levels of colon-derived solutes in hemodialysis patients. Clin J Am Soc Nephrol. 2014;9(9):1603–1610. doi: 10.2215/CJN.00490114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Vaziri ND, Liu SM, Lau WL, et al. High amylose resistant starch diet ameliorates oxidative stress, inflammation, and progression of chronic kidney disease. PLoS One. 2014;9(12):e114881. doi: 10.1371/journal.pone.0114881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Nakabayashi I, Nakamura M, Kawakami K, et al. Effects of synbiotic treatment on serum level of p-cresol in haemodialysis patients: a preliminary study. Nephrol Dial Transplant. 2011;26(3):1094–1098. doi: 10.1093/ndt/gfq624. [DOI] [PubMed] [Google Scholar]
- 145.Prakash S, Chang TM. Genetically engineered E. coli cells containing K. aerogenes gene, microencapsulated in artificial cells for urea and ammonia removal. Biomater Artif Cells Immobilization Biotechnol. 1993;21(5):629–636. doi: 10.3109/10731199309117387. [DOI] [PubMed] [Google Scholar]
- 146.Prakash S, Chang TM. Microencapsulated genetically engineered live E. coli DH5 cells administered orally to maintain normal plasma urea level in uremic rats. Nat Med. 1996;2(8):883–887. doi: 10.1038/nm0896-883. [DOI] [PubMed] [Google Scholar]
- 147.Schulman G, Agarwal R, Acharya M, Berl T, Blumenthal S, Kopyt N. A multicenter, randomized, double-blind, placebo-controlled, dose-ranging study of AST-120 (Kremezin) in patients with moderate to severe CKD. Am J Kidney Dis. 2006;47(4):565–577. doi: 10.1053/j.ajkd.2005.12.036. [DOI] [PubMed] [Google Scholar]
- 148.Vaziri ND, Yuan J, Khazaeli M, Masuda Y, Ichii H, Liu S. Oral activated charcoal adsorbent (AST-120) ameliorates chronic kidney disease-induced intestinal epithelial barrier disruption. Am J Nephrol. 2013;37(6):518–525. doi: 10.1159/000351171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Schulman G, Berl T, Beck GJ, et al. Randomized Placebo-Controlled EPPIC Trials of AST-120 in CKD. J Am Soc Nephrol. 2015;26(7):1732–1746. doi: 10.1681/ASN.2014010042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Rossi M, Johnson DW, Morrison M, et al. SYNbiotics Easing Renal failure by improving Gut microbiologY (SYNERGY): a protocol of placebo-controlled randomised cross-over trial. BMC Nephrol. 2014;15:106. doi: 10.1186/1471-2369-15-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Andriamihaja M, Lan A, Beaumont M, et al. The deleterious metabolic and genotoxic effects of the bacterial metabolite p-cresol on colonic epithelial cells. Free Radic Biol Med. 2015;85:219–227. doi: 10.1016/j.freeradbiomed.2015.04.004. [DOI] [PubMed] [Google Scholar]
- 152.Watanabe H, Miyamoto Y, Honda D, et al. p-Cresyl sulfate causes renal tubular cell damage by inducing oxidative stress by activation of NADPH oxidase. Kidney Int. 2013;83(4):582–592. doi: 10.1038/ki.2012.448. [DOI] [PubMed] [Google Scholar]
- 153.Strid H, Simren M, Stotzer PO, Ringstrom G, Abrahamsson H, Bjornsson ES. Patients with chronic renal failure have abnormal small intestinal motility and a high prevalence of small intestinal bacterial overgrowth. Digestion. 2003;67(3):129–137. doi: 10.1159/000071292. [DOI] [PubMed] [Google Scholar]
- 154.Simenhoff ML, Dunn SR, Zollner GP, et al. Biomodulation of the toxic and nutritional effects of small bowel bacterial overgrowth in end-stage kidney disease using freeze-dried Lactobacillus acidophilus. Miner Electrolyte Metab. 1996;22(1–3):92–96. [PubMed] [Google Scholar]
- 155.Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol. 2014;39(3):230–237. doi: 10.1159/000360010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Shi K, Wang F, Jiang H, et al. Gut bacterial translocation may aggravate microinflammation in hemodialysis patients. Dig Dis Sci. 2014;59(9):2109–2117. doi: 10.1007/s10620-014-3202-7. [DOI] [PubMed] [Google Scholar]
- 157.de Almeida Duarte JB, de Aguilar-Nascimento JE, Nascimento M, Nochi RJ., Jr Bacterial translocation in experimental uremia. Urol Res. 2004;32(4):266–270. doi: 10.1007/s00240-003-0381-7. [DOI] [PubMed] [Google Scholar]
- 158.Vaziri ND, Dure-Smith B, Miller R, Mirahmadi MK. Pathology of gastrointestinal tract in chronic hemodialysis patients: an autopsy study of 78 cases. Am J Gastroenterol. 1985;80(8):608–611. [PubMed] [Google Scholar]
- 159.Richardson AJ, McKain N, Wallace RJ. Ammonia production by human faecal bacteria, and the enumeration, isolation and characterization of bacteria capable of growth on peptides and amino acids. BMC Microbiol. 2013;13:6. doi: 10.1186/1471-2180-13-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Campbell RA, Grettie DP, Bartos F, Bartos D, Marton LJ. Uremic polyamine dysmetabolism. Proc Clin Dial Transplant Forum. 1978;8:194–198. [PubMed] [Google Scholar]
- 161.Igarashi K, Ueda S, Yoshida K, Kashiwagi K. Polyamines in renal failure. Amino Acids. 2006;31(4):477–483. doi: 10.1007/s00726-006-0264-7. [DOI] [PubMed] [Google Scholar]
- 162.Kushner D, Beckman B, Nguyen L, et al. Polyamines in the anemia of end-stage renal disease. Kidney Int. 1991;39(4):725–732. doi: 10.1038/ki.1991.88. [DOI] [PubMed] [Google Scholar]
- 163.Perna AF, Ingrosso D, Lombardi C, et al. Homocysteine in uremia. Am J Kidney Dis. 2003;41(3 Suppl 1):S123–S126. doi: 10.1053/ajkd.2003.50100. [DOI] [PubMed] [Google Scholar]
- 164.Turkmen K, Erdur FM. The relationship between colonization of Oxalobacter formigenes serum oxalic acid and endothelial dysfunction in hemodialysis patients: from impaired colon to impaired endothelium. Med Hypotheses. 2015;84(3):273–275. doi: 10.1016/j.mehy.2015.01.010. [DOI] [PubMed] [Google Scholar]
- 165.Cerini C, Dou L, Anfosso F, et al. P-cresol, a uremic retention solute, alters the endothelial barrier function in vitro. Thromb Haemost. 2004;92(1):140–150. doi: 10.1160/TH03-07-0491. [DOI] [PubMed] [Google Scholar]
- 166.Dou L, Bertrand E, Cerini C, et al. The uremic solutes p-cresol and indoxyl sulfate inhibit endothelial proliferation and wound repair. Kidney Int. 2004;65(2):442–451. doi: 10.1111/j.1523-1755.2004.00399.x. [DOI] [PubMed] [Google Scholar]
- 167.Nakabayashi I, Nakamura M, Kawakami K, et al. Effects of synbiotic treatment on serum level of p-cresol in haemodialysis patients: a preliminary study. Nephrol Dial Transplant. 2011;26(3):1094–1098. doi: 10.1093/ndt/gfq624. [DOI] [PubMed] [Google Scholar]
- 168.Evenepoel P, Bammens B, Verbeke K, Vanrenterghem Y. Acarbose treatment lowers generation and serum concentrations of the protein-bound solute p-cresol: a pilot study. Kidney Int. 2006;70(1):192–198. doi: 10.1038/sj.ki.5001523. [DOI] [PubMed] [Google Scholar]
- 169.The Effect of Arabinoxylan-oligosaccharides (AXOS) on Intestinal Generation of Microbial Metabolites in Chronic Kidney Disease. 2015 https://clinicaltrialsgov/ct2/show/study/NCT02141815?term=Bj%C3%B6rn+Meijers&rank=4.