Journal Information
Vol. 37. Issue. 1.January - February 2017
Pages 1-114
Visits
17922
Vol. 37. Issue. 1.January - February 2017
Pages 1-114
Review
Open Access
Gut microbiota in chronic kidney disease
Microbiota intestinal en la enfermedad renal crónica
Visits
17922
Secundino Cigarran Guldrisa,
Corresponding author
, Emilio González Parrab, Aleix Cases Amenósc
a Sección de Nefrología, Hospital Da Costa, Burela, Lugo, Spain
b Servicio de Nefrología, Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
c Servicio de Nefrología, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (5)
Table 1. Intestinal microflora in relation to perinatal events.4,5
Table 2. Gut microbiota changes in CKD.
Table 3. Effects of different uraemic toxins at the cellular and tissue level.
Table 4. Clinical studies with probiotics in patients with CKD and their effects.
Table 5. Clinical studies with symbiotics in patients with CKD and their effects.
Show moreShow less
Abstract

The intestinal microflora maintains a symbiotic relationship with the host under normal conditions, but its imbalance has recently been associated with several diseases.

In chronic kidney disease (CKD), dysbiotic intestinal microflora has been reported with an increase in pathogenic flora compared to symbiotic flora. An enhanced permeability of the intestinal barrier, allowing the passage of endotoxins and other bacterial products to the blood, has also been shown in CKD. By fermenting undigested products that reach the colon, the intestinal microflora produce indoles, phenols and amines, among others, that are absorbed by the host, accumulate in CKD and have harmful effects on the body. These gut-derived uraemic toxins and the increased permeability of the intestinal barrier in CKD have been associated with increased inflammation and oxidative stress and have been involved in various CKD-related complications, including cardiovascular disease, anaemia, mineral metabolism disorders or the progression of CKD. The use of prebiotics, probiotics or synbiotics, among other approaches, could improve the dysbiosis and/or the increased permeability of the intestinal barrier in CKD.

This article describes the situation of the intestinal microflora in CKD, the alteration of the intestinal barrier and its clinical consequences, the harmful effects of intestinal flora-derived uraemic toxins, and possible therapeutic options to improve this dysbiosis and reduce CKD-related complications.

Keywords:
Chronic kidney disease
Gut microbiota
Dysbiosis
Uraemic toxins
Inflammation
Resumen

La microflora intestinal mantiene una relación simbiótica con el huésped en condiciones normales, sin embargo, su alteración se ha asociado recientemente con numerosas enfermedades.

En la enfermedad renal crónica (ERC) se ha descrito una disbiosis en la microflora intestinal con un aumento de la flora patógena sobre la simbionte. Además, la permeabilidad de la barrera intestinal está aumentada, lo que permite el paso de endotoxinas y otros productos bacterianos a la sangre. La microflora intestinal, mediante la fermentación de productos no digeridos que alcanzan el colon, produce indoles, fenoles, o aminas, entre otros, que son absorbidos por el huésped, se acumulan en la ERC y tienen efectos deletéreos sobre el organismo. Estas toxinas urémicas generadas en el intestino y el aumento de la permeabilidad de la barrera intestinal en la ERC se han asociado a un aumento de la inflamación y el estrés oxidativo, y están implicados en diversas complicaciones asociadas a la ERC, como la enfermedad cardiovascular, la anemia, las alteraciones del metabolismo mineral o la progresión de la ERC. El uso de prebióticos, probióticos o simbióticos, entre otras aproximaciones, podrían mejorar la disbiosis o el aumento de la permeabilidad de la barrera intestinal en la ERC.

En este artículo se revisan la situación de la microflora intestinal en la ERC, la alteración de la barrera intestinal y sus consecuencias clínicas, los efectos deletéreos de las toxinas urémicas derivadas de la microflora intestinal, así como las posibles opciones terapéuticas para mejorar esta disbiosis y reducir las complicaciones de la ERC.

Palabras clave:
Enfermedad renal crónica
Microbiota intestinal
Disbiosis
Toxinas urémicas
Inflamación
Full Text
What are the microbiota and the microbiome?

Since Hippocrates (400 BC) established that “death lies in the intestines”, their influence on the health of human beings has been well known.

The germs that inhabit our body are called the microbiota, and their collective genomes, the microbiome. More than 100 trillion germs (1014) cohabit with us throughout our lives, representing 10 times the number of cells that make up our body and constituting 1.5–2kg of our weight.1,2 The concentration of germs in the digestive tract gradually increases from the stomach to the colon, where they reach the highest concentration (up to 1011 microorganisms/g of faeces) and diversity. The gut microbiota plays an important role in metabolic, nutritional, physiological, and immunological processes, and constitutes a true ecosystem.3 The human microbiome is our second genome, which has more than 3 million genes (100 times more genes than the human genome itself) and is the subject of research by the Human Microbiome Project Consortium.4–6

Originally, the gut microbiota is formed through the placenta, where low levels of non-pathogenic germs, especially the phyla firmicutes, bacteroidetes, and Fusobacteria are nested. In the first years of life, feeding, type of birth, hygiene, and use of antibiotics condition the formation of the intestinal microbiome.7,8 Different species of germs colonise and are originated during various events (Table 1).

Table 1.

Intestinal microflora in relation to perinatal events.4,5

Exposure  Intestinal flora 
Vaginal canal  Bifidobacterium, bacteroids, Lactobacillus, prevotella, enterococci, streptococci, Clostridiaeceae 
Post-caesarean delivery  Staphylococcus, Corynebacterium Propionibacterium. Lower amount of Bifidobacterium and bacteroides 
Breast-feeding  Bifidobacterium, bacteroides, Lactobacillus, clostridia, actinobacteria and firmicutes 
Artificial breastfeeding  Bacteroides, clostridia, Enterobacteriaceae 

The gut microbiota is established in the first 2–3 years of life as a dynamic ecosystem, dominated by bifidobacteria; their composition increases in richness and diversity until reaching their maximum complexity in adulthood, when the dominant species are bacteroidetes, firmicutes and actinobacteria.9–11

Bacterial communities that lie in the intestine are, therefore, a combination of different types and amounts of bacteria and 3 different groups of microbiota or enterotypes have been identified in humans.12 The phylogenetic composition of intestinal microflora tends to be similar between individuals from the same region, from the same family and also with a similar diet which plays a significant role in their composition.13,14

Dietary habits affect the composition of the gut microbiota. Since the microbiota is in contact with a significant number of neural cells and immunological cells, it directs the maturation of the immune system in childhood and contributes to the maintenance of its homeostasis during life.2 Complex polysaccharides, which are not digested by enzymes in the small intestine, are metabolised by the colon microflora. These polysaccharides are degraded and fermented in the large intestine and converted into short chain fatty acids (SCFAs) and gases (CO2 and H2). A high intestinal content of fructose promotes the formation of butyrate which is produced by bacteria. Dietary supplementation with specific polysaccharides may promote the growth of “healthy” germs (Bifidobacterium, Lactobacillus), the production of SCFA and it may decrease intestinal pH that inhibits the growth of pathogenic bacteria.15,16

In the ageing process, progressive damage occurs to the morphology and function of the different systems and the microbiota becomes less diverse and more dynamic, characterised by the predominance of bacteroidetes over firmicutes, with an increase of Protobacteria spp. and a decrease of Bifidobacterium. This is evidenced by rRNA techniques and it has been analysed as part of the study ELDERMET.17–19 Major changes in the microbiota are found in the colon of persons over 60 years of age. The significance of these changes is yet to be clarified.20

Faecal bacteria such as Escherichia coli, which divides every 20min, are genetically prepared to be highly adaptative, and always survive even though their host ages. Without this plasticity, we probably would not have been able to cope with changes in lifestyle and dietary habits, as evidenced by the transition from the Palaeolithic to the dietary habits of Western societies.21

Functionally, the gut microbiota provides nutrients and energy to the body through fermentation of nondigestible foods in the large intestine. The most important fermentation products deriving from the fermentation are the SCFA, which serve as a source of energy to intestinal cells and bacteria, and contribute to energy expenditure, satiety, and glucose homeostasis.22 Other relevant functions of the gut microbiota are the endogenous synthesis of certain vitamins and amino acids, the metabolism of bile acids, or the maintenance of the integrity of intestinal barriers, which protect the host from pathogenic germs.

Thus, the gut microbiota is involved in the maturation of the immune system in infancy and contributes to the maintenance of its homeostasis throughout life.23

Gut microbiota in chronic kidney disease

From the early stages of chronic kidney disease (CKD) there is a quantitative and qualitative alteration of intestinal microflora (dysbiosis); so the composition and metabolic activities of microflora are changed in CKD and this is a hot and innovative topic in nephrology literature. These alterations include changes in intestinal transit, decreased protein absorption, decrease in dietary fibre intake, treatment with oral iron and frequent use of antibiotics.

All of this contributes to systemic inflammation and the accumulation of uraemic toxins that are absorbed by intestine and eliminated by the kidney. Inflammation and uraemic toxins play a central role in the pathophysiology of atherosclerosis, as well as in other complications associated with CKD24–27; that will be reviewed below.

Patients with CKD are polymedicated. Some drugs frequently prescribed to these patients may alter intestinal microflora, especially antibiotics,28,29 but others may also slow intestinal transit, phosphorus binders, ion exchange resins,30 or iron supplements that may have an effect on microflora but it is not well defined.31,32

Change in intestinal barriers in chronic kidney disease

Changes in intestinal barriers with an increased intestinal permeability is common in CKD (Table 2).

Table 2.

Gut microbiota changes in CKD.

Intestinal tract  Normal  CKD/ACKD 
Stomach  Lactobacillus  No change 
  Helicobacter   
Duodenum  Staphylococcus  Increased 
  Streptococcus   
  Lactococcus   
Jejunum  Enterococcus  Increased 
  Streptococcus   
  Lactobacillus   
Ileum  Enterobacteriaceae  Increased 
  Bacteroides   
  Clostridium   
  Bacterial fragments   
Colon  Firmicutes  Increased: 
  Bacteroides  Proteobacteria, 
  Actinobacteria  Enterobacteria, E. coli
  Proteus  Acinetobacter, Proteus spp. 
  Clostridium
Lactobacilli 
Decreased: 
  Prevotellaceae  Lactobacillus, Bifidobacterium spp. 
  Fusobacterium  Aerobic overgrowth of at least 100 times 
  TM7  Increase of Clostridium perfringens 

CKD, chronic kidney disease; ACKD, advanced chronic kidney disease.

Increasing urea levels and expanding bacteria with urease causes an increase ammonium production in the intestinal lumen and induce changes in intestinal pH that produces an alteration of intestinal permeability by affecting the tight junctions of the enterocyte. Vaziri et al. have demonstrated a marked reduction of tight junction proteins, claudin-1, occludin, and ZO1, in the colonic mucosa in CKD; this is associated with an infiltration of mononuclear leukocytes in the lamina propria and a marked thickening of the colon wall.33 There is histological evidence of chronic inflammation of the intestinal tract including oesophagitis, gastritis, etc.33,34

The presence of frequent oedema and hypervolaemia in CKD may aggravate intestinal barrier dysfunction in CKD patients on, haemodialysis, or peritoneal dialysis. In addition, excessive ultrafiltration and episodes of hypotension during haemodialysis may cause episodes of transient intestinal ischaemia which increases the permeability of the intestinal barrier facilitating the passage of endotoxins.33

In renal transplant patients, the investigation of the gut microbiota is in its infancy. It is known that inflammatory processes, such as graft ischaemia time, baseline disease and immunosuppressive drugs may play a relevant role in the alteration of the intestinal barrier.35,36

Intestinal microflora as a cause of inflammation in chronic kidney disease

In CKD, the decreased clearance of proinflammatory cytokines, is associated with the development of oxidative stress and inflammation. The later are contributing factors to the progression of the disease and its complications, including cardiovascular disease, cachexia, and anaemia. Oxidative stress and chronic inflammation stimulate the NF-κB transcription factor, which is the key regulator of proinflammatory cytokines and chemokines. Increased permeability of intestinal barriers in CKD patients favours the translocation of bacterial products of intestinal origin, as evidenced by the presence of DNA fragments of circulating intestinal pathogens (aerobic and anaerobic), both in patients in different stages of CKD and on renal replacement therapy.37–39 The increase in circulating bacterial products of intestinal origin activates innate immunity, promotes the inflammatory state associated with CKD and, increases the incidence of cardiovascular disease and mortality.40–42

Microbiota and uraemic toxins derived from the intestine in chronic kidney diseaseIntestinal production of uraemic toxins

The origin of uraemic toxins in CKD is multiple. The importance of toxins generated by intestinal microbial metabolism43 is increasingly recognised. Approximately 10g of proteins reach the colon daily, where they are degraded by intestinal bacteria to metabolites such as ammonium, amines, thiols, phenols and indoles. These colon fermentation products are eliminated through faeces, although a portion is absorbed and eliminated by the kidney, so these are accumulated in CKD.44 In CKD, the uraemic toxins derived from intestinal microflora are:

Phenols and indoles: p-cresol and indoxyl sulfate. Phenols include p-cresol, p-cresyl sulfate (PCS), p-cresyl glucuronide, phenylacetic acid, phenyl sulfate and phenol.45

  • -

    p-Cresol/p-cresyl sulfate: products of phenylalanine and tyrosine metabolism of intestinal anaerobic bacteria. p-Cresol is conjugated in the intestinal wall to PCS and p-cresyl glucuronide in the liver. PCS is the main circulating metabolite of p-cresol.46

  • -

    Phenol: mainly derives from ingestion, from the catabolism of tyrosine by intestinal bacteria, as well as from tobacco consumption.

  • -

    Phenylacetic acid: is the result of the degradation of phenylalanine.

Among the indoles are indoxyl sulfate (IS) and indoleacetic acid.45 Both originate from the degradation of tryptophan by intestinal bacteria and are subsequently sulfated in the liver into IS. Indoles and phenols are uraemic toxins bound to proteins.47

Amines and polyamines: amines and polyamines are generated from intestinal microbial metabolism. An amino acid that is clinically relevant and of growing interest is trimethylamine N-oxide (TMAO). TMAO is produced by the intestinal metabolism of quaternary amines, such as choline/phosphatidylcholine, betaine, or l-carnitine. l-Carnitine, which is present in red meat, also induces the formation of TMAO and it is associated with an increase in cardiovascular disease.48 Dietary sources of TMAO are red meats, meats in general, egg yolks, liver, dairy products and saltwater fish. In CKD, TMAO accumulates and its levels depends on the glomerular filtration, but its binding to proteins is low, and it is well eliminated with dialysis.

Polyamines are organic cations including cadaverine, spermine, spermidine and putrescine. They come from the decarboxylation of l-arginine, l-ornithine or lysine in the intestine. In CKD patients, putrescine, spermidine, and spermine are increased in serum.49 These molecules have been shown to interact with insulin and lipoproteins, and contribute to the acceleration of atherosclerosis along with other factors such as hypertriglyceridemia.50

Biological and clinical consequences of the accumulation of uraemic toxins

The aforementioned uraemic toxins have been associated with deleterious biological effects in different tissues and cell lines51,52 (Table 3), and with an increased risk of the progression of CKD, morbidity and mortality.

  • a)

    Progression of CKD: both IS and PCS are associated to the development of fibrosis, deterioration of renal function and disease progression.52,53 In vitro studies have shown a deleterious effects of these molecules on renal tubular cells.54 In a prospective study in patients with stage 1–5 CKD, the predictive role of both molecules in disease progression was confirmed.55 In experimental animals, a diet rich in choline or TMAO preduces progressive tubulointerstitial fibrosis and renal dysfunction.56

  • b)

    Cardiovascular complications: in CKD patients, IS is associated with endothelial damage, arterial stiffness and aortic calcification57; and, in hemodialysis patients it is associated with atherosclerosis58 and endothelial dysfunction,59 it has a cardiac profibrotic effect, favours hypertrophy of myocardiocytes60 and it is a predisposing factor of atrial fibrillation.61 Similar vascular effects have been described with,62 which is a predictor of cardiovascular risk in CKD patients.63,64 In hemodialysis patients PCS and IS have been associated with peripheral vascular disease and thrombosis of vascular access.65 A recent meta-analysis confirms the relationship of these molecules to cardiovascular risk in CKD.66

    Likewise, indoleacetic acid is associated with oxidative stress and inflammation markers, and it is a predictor of mortality and cardiovascular events in CKD.67

    Elevated levels of TMAO predicts coronary atherosclerotic burden56 and mortality in patients with CKD,68,69 although this is not shown in all reports.70

  • c)

    Anaemia: IS has been associated with anaemia of the renal patient; it interfere with the adequate production of erythropoietin71,72 and increased eryptosis (programmed cell death of red blood cells).73 Polyamines are associated to anaemia in renal patients, through an intra-erythrocytic effect,74 reduces erythropoiesis, and inhibit the activity of erythropoietin.

  • d)

    Alterations of bone-mineral metabolism: IS reduces bone formation by promoting oxidative stress in osteoblasts and inducing resistance to PTH, which favours the development adynamic bone.75 There is a positive correlation between FGF-23 and IS serum levels, suggesting an association between this molecule and metabolic bone disease in uraemic patients.76 Likewise, less bone remodelling has been observed in uraemic rats with higher IS after a parathyroidectomy.77

  • e)

    Insulin resistance: In CKD patients the catabolism of insulin is reduced and often, they also have insulin resistance, which is associated with an increased risk of mortality; it seems that insulin resistance is related to some of the uraemic toxins.78

Table 3.

Effects of different uraemic toxins at the cellular and tissue level.

Organ/tissue  Toxin  Effect 
Endothelium  IS  Increased senescence 
    Induction of ROS and decrease in NO production 
  IS and PCS  Increased expression of ICAM-1, MCP-1 and tissue factor 
    Increased adhesion of leukocytes to the endothelium 
  PCS  Inhibition of proliferation, viability, and repair 
  IAA  Increased release of endothelial microparticles 
    Increased endothelial permeability 
    Increased ROS and inflammation and tissue factor expression 
    Apoptosis of endothelial cell progenitors 
Vascular smooth muscle fibre  IS  Increased proliferation 
    Increased production of tissue factor 
  PAA  Increased ROS and expression of osteoblastic proteins 
    Increased ROS production 
Vessels  IS  Increased aortic calcification and stiffness, expression of osteoblastic markers and OAT 
  IS and PCS  Increased cellular senescence 
  TMAO  Increased rolling and adhesion of leukocytes to the vessel 
    Accelerated atherosclerosis 
Leukocytes  PCS  Activation of the oxidative burst 
  IS  Increased adhesion to the endothelium 
  IS and IAA  Increased expression of mononuclear cell tissue factor 
  TMAO  Increased expression of scavenger receptors in macrophages 
Cardiac cells  IS  Hypertrophy of cardiomyocytes, production of collagen by myofibroblasts and inflammation 
Heart  IS  Myocardial hypertrophy, cardiac fibrosis and oxidative stress 
Renal tubular cells  PCS and IS  Activation of RAS, mesenchymal epithelial transition, and fibrosis 
    Increased expression of proinflammatory genes and cytokines 
  PCS  Increased methylation of the klotho gene and fibrosis 
  IS  Increased tubular damage 
  IS and IAA  Increased expression of MCP-1, ICAM-1, TGF-β and Smad3 
    Increased oxidative stress, inhibition of proliferation, increased expression of PAI-1 and NF-κB activation 
  PCS, IS, IAA  Decreased cell viability 
Kidneys  IS  Increased fibrosis and angiotensinogen expression 
    Decreased klotho expression and increased senescence 
  IAA  Increased glomerulosclerosis 
  TMAO  Increased monocyte infiltration 
    Increased glomerular sclerosis and interstitial fibrosis 
    Increased tubulointerstitial fibrosis and collagen deposition 
Adipocytes  PCS and IS  Increased insulin resistance 
Osteoclasts  IS  Alteration of differentiation and function 
Osteoblasts  PCS and IS  Decreased cell viability and cell proliferation and increased ROS production 
  IS  Decreases PTH receptor expression 
    Promotes apoptosis 
  PAA  Inhibits proliferation and differentiation 

PAA, phenylacetic acid; IAA, indole acetic acid; IS, indoxyl sulfate; OAT, organic acid transporters; PC, p-cresol; PCS, p-cresyl sulfate; PTH, parathyroid hormone; RAS, renin–angiotensin system; ROS, oxygen free radicals; TMAO, trimethylamine N-oxide.

Source: Modified and extended from Biagi et al.17
Prevention and treatment of dysbiosis

In recent years there is a growing interest in restoring the symbiosis of intestinal microflora in CKD aiming to reduce the generation of uraemic toxins, oxidative stress, and inflammation.79

  • a)

    High fibre diet: a high fibre diet increases the production of SCFA, which provides energy to the intestinal flora and allows amino acids that reach the colon to be incorporated into bacterial proteins and be excreted instead of being fermented into uraemic solutes. In addition, SCFAs are used as substrate by the intestinal mucosa helping to maintain their functionality and integrity. Fibre increases intestinal transit reducing the time for fermentation of amino acids and improves the composition of microflora which reduces the production of undesirable solutes. In CKD patients, there is a direct relationship between dietary protein/fibre ratio and PCS and IS levels, so a diet with a low protein/fibre ratio should be beneficial.80 In healthy subjects, a vegetarian diet, as compared with the omnivore diet, reduces the generation IS or PCS; this effect was related to the higher fibre and lower protein content of the vegetarian diet.81 A very low protein diet (0.3g/kg body weight/day) supplemented with amino acid keto-analogues also reduces IS levels in patients with CKD.82

    Several therapeutic interventions have recently explored to improve the dysbiosis of the intestinal microflora, reduce the absorption of uraemic toxins and the passage of endotoxins from the intestinal lumen.

  • b)

    Prebiotics, probiotics, and symbiotics: the generation of uraemic toxins could be reduced by selectively increasing saccharolyticbacteria (which digest dietary fibre) and decreasing proteolytic bacteria (protein and amino acid fermenters) in the colon. The main regulator of metabolism of colon bacteria is the availability of nutrients and specifically the rate of fermentable carbohydrates vs. nitrogen.

    Prebiotics are non-digestible food components which, through selective fermentation, allow for specific changes in the composition or activity in gastrointestinal microflora, which are beneficial to the health and well-being of the host. Prebiotics stimulate the growth or activity of one or a limited number of bacteria in the colon; they may increase carbohydrate fermentables vs. nitrogen; they include inulin, fructooligosaccharides, galactooligosaccharides, etc. Inulin enriched with oligofructose reduces the generation of PCS and the serum concentrations in hemodialysis patients, but has no effect on IS.83 Resistant starch reduces IS levels in hemodialysis patients and reduces PCS but not significantly.84 In a CKD rat model, a diet rich in resistant starch delayed the progression of CKD and attenuated oxidative stress and inflammation.85 Currently, a randomised, crossover, double-blind, phase 2 clinical trial in patients with stage 3b-4 CKD is examining the effect of the supplementation of arabinoxylan-oligosaccharides on plasma levels of PCS and indole derivatives urinary excretion of these compounds and insulin resistance are also examined.86

    Probiotics are defined as “living micro-organisms” that, being administered in adequate amounts, provide a health benefit to the host. A recent review evaluates the potential benefits of probiotics in general and especially in CKD.87 The efficacy of probiotics to decrease levels of uraemic toxins and to delay the progression of CKD has been investigated in in vitro models, animal models and in patients with CKD. However, to date, there are no large-scale quality intervention studies and studies on clinical events to support their widespread use. There are only small studies, most of which,88,92–95 but not all,97 observe a decrease in uraemic toxin levels. Administration of Bifidobacterium longum in enteric capsules to patients with CKD had minimal effects on the progression of the disease in patients with CKD.91 However, a randomised, double-blind trial in patients on peritoneal dialysis observed a significant reduction in serum proinflammatory endotoxin and cytokine levels, an increase in serum IL-10 levels, and the preservation of residual renal function after 6 months of treatment with a probiotic96 (Table 4).

    Table 4.

    Clinical studies with probiotics in patients with CKD and their effects.

    Author and year  Probiotic  Type of study  Results 
    Hida et al., 199688  Lebenin  Observational, patients in HD (n=25), 4 weeks  ↓ Indicate in faeces and serum
    ↓ p-Cresol in faeces 
    Simenhoff et al., 199689  Lactobacillus acidophilus  Observational, patients in HD (n=8)  ↓ Dimethylamine
    ↓ Nitrosodimethylamine 
    Takayama et al., 200390  Bifidobacterium longum JCM008  Non-randomised, placebo-controlled. Patients in HD (n=22), 5 weeks  ↓ Indoxyl sulfate 
    Ando et al., 200391  Bifidobacterium longum  Observational, patients with CKD (n=27), 6 months  Reduction of the progression of CKD in patients with con Cr ≥4mg/dl or P4mg/dl 
    Taki et al., 200592  Bifidobacterium longum  Non-randomised, placebo-controlled. Patients in HD (n=27), 12 weeks  ↓ Indoxyl sulfate, homocysteine and triglycerides 
    Ranganathan et al., 200993  Lactobacillus acidophilus KB31, Streptococcus thermophilus KB27, Bifidobacterium longum KB35  Randomised, double-blind, crossover, placebo-controlled. Patients with CKD 3–4 (n=16), 6 months  BUN
    Uric acid
    ↑ Quality of life 
    Ranganathan et al., 201094  Lactobacillus acidophilus KB31, Streptococcus thermophilus KB27, Bifidobacterium longum KB35  Multicentre, randomised, double-blind, placebo-controlled. Patients with CKD 3–4 (n=46), 6 months  BUN
    ↑ Quality of life
    Insurance 
    Miranda Alatriste et al., 201495  Lactobacillus casei shirota  Randomised, placebo-controlled, patients with CKD 3–4 (n=30), 8 weeks  ↓ Urea 
    Wang et al., 201596  Bifidobacterium bifidum A218, Bifidobacterium catenulatum A302, Bifidobacterium longum A101, Lactobacillus plantarum A87  Randomised, double-blind, placebo-controlled, patients in PD (n=39), 6 months  ↓ TNF-α, IL-5, IL-6 and endotoxin
    ↑ IL-10
    Residual preservation of kidney function 
    Natarajan et al., 201497  Renadyl  Randomised, double-blind, placebo-controlled, patients in HD (n=22), 8 weeks  No changes in quality of life
    Tendency towards reduction of indoxyl glucuronide, CRP and leucocyte count 

    Cr, creatinine; PD, peritoneal dialysis; CKD, chronic kidney disease; HD, haemodialysis; IL, interleukin; P, phosphorus; CRP, C-reactive protein.

    Symbiotics are probiotic supplements combined with prebiotics. In hemodialysis patients, on treatment with a symbiotic there is a decrease in the level of PCS, but not those of IS,98 which was confirmed in another study.102 Another study observed a delay in the progression of CKD with symbiotic treatment,100 while another study did not observe a significant improvement in inflammation markers.103 Finally, a randomised, double-blind, crossover study in patients with CKD104 demonstrated a reduction in PCS levels, a non-significant decrease in IS and an increase in bifidobacteria and reduction of faecal ruminococcus, but no change in inflammation markers, oxidative stress, or endotoxins, although a slight increase in albuminuria was observed (Table 5).

    Table 5.

    Clinical studies with symbiotics in patients with CKD and their effects.

    Author and year  Symbiotic  Study  Results 
    Nakabayashi et al., 201098  Lactobacillus casei shirota, Bifidobacterium breve yakult and galactooligosaccharides  Observational, patients in HD (n=9), 4 weeks  ↓ p-Cresol in plasma
    Normalisation of bowel habits
    Association of p-cresol and constipation 
    Ogawa et al., 201299  Bifidobacterium longum JBL01 and oligosaccharides  Observational, patients in HD (n=15). Control group patients in HD (n=16), 4 weeks  ↓ P levels that returned to baseline 2 weeks later 
    Pavan et al., 2014100  Probiotic and prebiotic  Prospective, open, randomised, placebo-controlled. Patients with CKD 3–5 (n=24), 12 months  Reduction in the progression of CKD 
    Cruz-Mora et al., 2014101  Lactobacillus acidophilus, Bifidobacterium lactis and inulin  Double-blind, randomised, placebo-controlled. Patients in HD (n=18), 2 months  ↑ Bifidobacteria in faeces
    Lactobacilli in faeces (in the 2 groups)
    Improvements of GI symptoms 
    Guida et al., 2014102  Lactobacillus plantarum, Lactobacillus casei subsp. Rhamnosus, Lactobacillus gasseri, Bifidobacterium infantis, Bifidobacterium longum, Lactobacillus acidophilus, Lactobacillus salivarius, Lactobacillus sporogenes and Streptococcus thermophilus inulin and resistant tapioca starch  Randomised, double-blind, placebo-controlled. Patients with CKD 3–4 (n=30), 4 weeks  ↓ p-Cresol in plasma
    With no changes in GI symptoms 
    Viramontes-Hörner et al., 2015103  Lactobacillus acidophilus and Bifidobacterium lactis+prebiotic (inulin)  Randomised, double-blind, placebo-controlled. Patients in HD (n=42), 2 months  Tendency to diminish levels of CRP and TNF-α. Improvement of GI symptoms 
    Rossi et al., 2014104  Lactobacillus, bifidobacteria and Streptococcus generates+inulin, fructooligosaccharides and galactooligosaccharides  Randomised, double-blind, crossover, placebo-controlled. Patients with CKD 4–5, 6 weeks with a washout of 4 weeks  Decreased PCS, decreased NS of IS
    Increased bifidobacteria and decreased ruminococcaceae in faeces. No changes in oxidative stress or inflammation markers and slight increase in albuminuria 

    CRD, chronic kidney disease; GI, gastrointestinal; HD, haemodialysis; NO, nonsignificant; P, phosphorus; CRP, C-reactive protein; PCS, p-cresyl sulfate.

    One of the major limitations of probiotic or symbiotic therapy is that no study has yet demonstrated the sustained survival of probiotics in the dysbiotic colon of patients with CKD. There are also no studies that have evaluated the effect of these treatments on the levels of TMAO in this population. In choosing probiotics, the contribution of urease-containing bacteria must be considered, since they may increase intestinal ammonia generation, which may damage epithelial tight junctions, and increase intestinal permeability to the passage of endotoxins from the intestinal lumen.33,34

  • c)

    Adsorptive therapies: the use of oral sorbents could decrease uraemic toxins and circulating intestinal endotoxins. AST-120 oral sorbents decrease IS levels in a dose-dependent manner.105 In addition, a reduction in the IS, PCS, or phenyl sulfate and oxidative stress levels have been described in patients on haemodialysis.106 Other authors have reported that the administration of AST-120 improves the erythropoietic response to CERA.107 AST-120 improves intestinal barrier dysfunction and decreases endotoxin plasma levels, inflammation markers and oxidative stress in a CKD model in rats.108

Although small randomised, controlled studies in experimental animals and retrospective studies in patients have indicated a nephroprotective effect of AST-120 (reviewed by Schulman et al.109), a subsequent large randomised, controlled trial in patients with CKD was unable to confirm this.109 The study had some methodological limitations, but also suggested the possibility that the objective of treating specific uraemic toxins may not be sufficient. However, another retrospective study of the long-term effects of AST-120 on patients with stage 3–5 CKD showed a reduction in the risk of progression to dialysis, mortality, cardiac events and vascular accident vs. those patients who did not receive it.110

Although a beneficial effect of sevelamer on IS and PCS has been described in in vitro studies, in vivo studies in mice or patients have not demonstrated a reduction in the levels of these uraemic toxins.111 However, sevelamer does reduce endotoxin levels and systemic inflammation in patients on haemodialysis.112,113

Key concepts

  • 1.

    In CKD, there is a dysbiosis of the intestinal microflora.

  • 2.

    Intestinal microflora generate uraemic toxins that are absorbed and accumulate in CKD, and are associated with increased oxidative stress and inflammation.

  • 3.

    In CKD, there is an increase in the permeability of the intestinal barrier that allows the passage into the systemic circulation of endotoxins and other bacterial products that aggravate the inflammatory state of CKD.

  • 4.

    Changes in diet composition could improve microflora dysbiosis in CKD, reduce uraemic toxin levels, or restore intestinal mucosal permeability in CKD patients.

  • 5.

    The use of probiotics, prebiotics or symbiotics opens an alternative in the treatment of intestinal dysbiosis associated with CKD, and may play a role in slowing the progression of CKD and in preventing relevant associated complications such as mortality and cardiovascular risk.

Conflicts of interest

The authors have no conflicts of interest to declare.

Acknowledgements

This work was supported by grants from Fondo de Investigaciones Sanitarias (PI16/01298), Madrid Society of Nephrology and REDinREN.

References
[1]
C. Palmer, E.M. Bick, D.B. DiGulio, D.A. Relman, P.O. Brown.
Development of the human infant intestinal microbiota.
[2]
R. Tojo, A. Suarez, M.G. Clemente, C. de los Reyes Gavilán, A. Margolles, M. Gueimonde, et al.
Intestinal microbiota in health and disease: role of bifidobacteria in gut homeostasis.
World J Gastroenterol, 20 (2014), pp. 15163-15176
[3]
A.M. O’Hara, F. Shanahan.
The gut flora as a forgotten organ.
EMBO Rep, 7 (2006), pp. 688-693
[4]
S. Hacquard, R. Garrido-Oter, A. Gonzalez, S. Spaepen, G. Ackermann, S. Lebeis, et al.
Microbiota and host nutrition across plant and animal kingdoms.
Cell Host Microbe, 17 (2015), pp. 603-616
[5]
C. Huttenhower, D. Gevers, R. Knight, S. Abubucker, J.H. Badger, on behalf of the Human Microbiome Project Consortium.
Structure, function and diversity of the healthy human microbiome.
Nature, 486 (2013), pp. 207-2014
[6]
D. Gevers, R. Knight, J.F. Petrosino, K. Huang, A.L. McGuire, B.W. Birren, et al.
The human microbiome project: a community resource for the healthy human microbiome.
PLoS Biol, 10 (2012), pp. e1001377
[7]
S.B. Meropol, A. Edwards.
Development of the infant intestinal microbiome: a bird's eye view of a complex process.
Birth Defects Res, 105 (2015), pp. 228-239
[8]
T.M. Marques, R. Wall, R.P. Ross, G.F. Fitzgerald, C.A. Ryan, C. Stanton.
Programming infant gut microbiota: influence of dietary and environmental factors.
Curr Opin Biotechnol, 21 (2010), pp. 149-156
[9]
P.A. Scholtens, R. Oozeer, R. Martin, K.B. Amor, J. Kno.
The early settlers: intestinal micro-biology in early life.
Annu Rev Food Sci Technol, 3 (2012), pp. 425-447
[10]
J.E. Koenig, A. Spor, N. Scalfone, A.D. Fricker, J. Stombaugh, R. Knight, et al.
Succession of microbial consortia in the developing infant gut microbiome.
Proc Natl Acad Sci U S A, 108 (2011), pp. 4578-4585
[11]
M. Rajilic-Stojanovic, H.G. Heilig, D. Molenaar, K. Kajander, A. Surakka, H. Smidt, et al.
Development and application of the human intestinal tract chip, a phylogenetic microarray: analysis of universally conserved phylotypes in the abundant microbiota of young and elderly adults.
Environ Microbiol, 11 (2009), pp. 1736-1751
[12]
N. Ottman, H. Smidt, W.M. de Vos, C. Belzer.
The function of our microbiota: who is out there and what they do?.
Front Cell Infect Microbiol, 2 (2012), pp. 104
[13]
M. Arumugam, J. Raes, E. Pelletier, D. le Paslier, T. Yamada, D.R. Mende, et al.
Enterotypes of the human gut microbiome.
Nature, 473 (2011), pp. 174-180
[14]
M.J. Claesson, I.B. Jeffery, S. Conde, S.E. Power, E.M. O’Connor, S. Cusack, et al.
Gut microbiota composition correlates with diet and health in the elderly.
Nature, 488 (2012), pp. 178-184
[15]
J.E. Suarez.
Microbiota autóctona, probióticos y prebióticos.
[16]
C. Chassard, C. Lacroix.
Carbohydrates and the human microbiota.
Curr Opin Clin Nutr Metab Care, 16 (2013), pp. 453-460
[17]
E. Biagi, L. Nylund, M. Candela, R. Ostan, L. Bucci, E. Pini, et al.
Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians.
[18]
A.L. Man, E. Bertelli, S. Rentini, M. Regoli, G. Briars, M. Marini, et al.
Age-associated modifications of intestinal permeability and innate immunity in human small intestine.
Clin Sci (Lond), 129 (2015), pp. 515-527
[19]
S. Soenen, C.K. Rayner, K.L. Jones, M. Horowitz.
The ageing gastrointestinal tract.
Curr Opin Clin Metab Care, 19 (2016), pp. 12-18
[20]
S.C. Bischoff.
Microbiota and aging.
Curr Opin Clin Metab Care, 19 (2016), pp. 26-30
[21]
S. Quercia, M. Candela, C. Giuliani, S. Turroni, D. Luiselli, S. Rampelli, et al.
From lifetime to evolution: timescales of human gut microbiota adaptation.
Front Microbiol, 5 (2014), pp. 587
[22]
P.D. Cani, A. Everard, T. Duparc.
Gut microbiota, enteroendocrine functions and metabolism.
Curr Opin Pharmacol, 13 (2013), pp. 935-940
[23]
A. Hevia, S. Delgado, B. Sánchez, A. Margolles.
Molecular players involved in the interaction between beneficial bacteria and the immune system.
Front Microbiol, 6 (2015), pp. 1285
[24]
J.A. Aron-Wisnewsky, K. Clement.
The gut microbiome, diet, and links to cardiometabolic and chronic disorders.
Nat Rev Nephrol, 12 (2016), pp. 169-181
Epub 2015 Nov 30
[25]
D. Mafra, J.C. Lobo, F. Barros, L. Koppe, N. DVarizi, D. Fouque.
Role of altered intestinal microbiota in systemic inflammation and cardiovascular disease in chronic kidney disease.
Future Microbiol, 9 (2014), pp. 399-410
[26]
A. Sabatino, G. Regolisti, I. Brusasco, A. Cabassi, S. Morabito, E. Fiaccadori.
Alterations of intestinal barrier and microbiota in chronic kidney disease.
Nephrol Dial Transplant, 30 (2015), pp. 924-933
[27]
H.J. Anders, K. Andersen, B. Stecher.
The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease.
Kidney Int, 83 (2013), pp. 1010-1016
[28]
C. Jemberg, S. Löfmark, C. Edlund, J.K. Jansson.
Long-term impact of antibiotic exposure on the human intestinal microbiota.
Microbiology, 156 (2010), pp. 3216-3223
[29]
H.E. Jakobsson, C. Jemberg, A.F. Andersson, M. Sjölund-Karlsson, J.K. Jansson, L. Engstrand.
Short-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome.
[30]
M.J. Wu, C.S. Chang, C.H. Cheng, C.H. Chen, W.C. Lee, Y.H. Hsu, et al.
Colonic transit time in long-term dialysis patients.
Am J Kidney Dis, 44 (2004), pp. 322-327
[31]
G.A. Kortman, M. Raffatellu, D.W. Swinkels, H. Tjalsma.
Nutritional iron turned inside out: intestinal stress from a gut microbial perspective.
FEMS Microbiol Rev, 38 (2014), pp. 1202-1234
[32]
G. Weiss.
Dietary iron supplementation: a proinflammatory attack on the intestine?.
[33]
N.D. Vaziri.
CKD impairs barrier function and alters microbial flora of the intestine: a major link to inflammation and uremic toxicity.
Curr Opin Nephrol Hypertens, 21 (2012), pp. 587-592
[34]
S. Goncalves, R. Pecoits-Filho, S. Perreto, S.H. Barberato, A.E. Stinghen, E.G. Lima, et al.
Associations between renal function, volume status and endotoxemia in chronic kidney disease patients.
Nephrol Dial Transpl, 21 (2006), pp. 2178-2794
[35]
M.L. Alegre, R.B. Mannon, P.J. Mannon.
Thee microbiota, the immune system and the allograft.
Am J Transplant, 14 (2014), pp. 1236-1248
[36]
J.S. Bromberg, W.F. Fricke, C.C. Brinkman, T. Simon, E.F. Mongodin.
Microbiota – implications for immunity and transplantation.
Nat Rev Nephrol, 11 (2015), pp. 342-353
[37]
K. Shi, F. Wang, H. Jiang, H. Liu, M. Wei, Z. Wang, et al.
Gut bacterial translocation may aggravate microinflammation in hemodialysis patients.
Dig Dis Sci, 59 (2014), pp. 2109-2117
[38]
F. Wang, H. Jiang, K. Shi, Y. Ren, P. Zhan, S. Cheng.
Gut bacterial translocation is associated with microinflammation in end-stage renal disease patients.
Nephrology (Carlton), 17 (2012), pp. 733-738
[39]
D. Mafra, D. Fouque.
Gut microbiota and inflammation in chronic kidney disease patients.
Clin Kidney J, 8 (2015), pp. 332-334
[40]
C.W. McIntyre, L.E. Harrison, M.T. Eldehni, H.J. Jefferies, C.C. Szeto, S.G. John, et al.
Circulating endotoxemia: a novel factor in systemic inflammation and cardiovascular disease in chronic kidney disease.
Clin J Am Soc Nephrol, 6 (2011), pp. 133-141
[41]
R. Vanholder, G. Glorieux.
The intestine and the kidneys: a bad marriage can be hazardous.
Clin Kidney J, (2015), pp. 168-179
[42]
A. Ramenazani, Z.A. Massy, B. Meijers, P. Evenepoel, R. Vanholder, D.S. Raj.
The gut microbiome in uremia: potential therapeutic target.
Am J Kidney Dis, 67 (2016), pp. 483-498
Epub 2015 Nov 15
[43]
W.R. Wikoff, A.T. Anfora, J. Liu, P.G. Schultz, S.A. Lesley, E.C. Peters, et al.
Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites.
Proc Natl Acad Sci U S A, 106 (2009), pp. 3698-3703
[44]
P. Evenepoel, B.K. Meijers, B.R. Bammens, K. Verbeke.
Uremic toxins originating from colonic microbial metabolism.
Kidney Int Suppl, (2009), pp. S12-S19
[45]
N. Jourde-Chiche, L. Dou, C. Cerini, F. Dignat-George, R. Vanholder, P. Brunet.
Protein-bound toxins – update.
Semin Dial, 22 (2009), pp. 334-339
[46]
A.W. Martinez, N.S. Recht, T.H. Hostetter, T.W. Meyer.
Removal of P-cresol sulfate by hemodialysis.
J Am Soc Nephrol, 16 (2005), pp. 3430-3436
[47]
R. Vanholder, R. de Smet, G. Glorieux, A. Argilés, U. Baurmeister, P. Brunet, European Uremic Toxin Work Group (EUTox), et al.
Review on uremic toxins: classification, concentration, and interindividual variability.
Kidney Int, 63 (2003), pp. 1934-1943
[48]
R.A. Koeth, Z. Wang, B.S. Levison, J.A. Buffa, E. Org, B.T. Sheehy, et al.
Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis.
Nat Med, 19 (2013), pp. 576-585
[49]
A. Saito, T. Takagi, T.G. Chung, K. Ohta.
Serum levels of polyamines in patients with chronic renal failure.
Kidney Int, 24 (1983), pp. S2-S4
[50]
W. Lutz.
A uremic peptide containing polyamine: formation and possible role in uremic hypertriglyceridemia.
Physiol Chem Phys, 12 (1980), pp. 451-456
[51]
R. Vanholder, E. Schepers, A. Pletinck, E.V. Nagler, G. Glorieux.
The uremic toxicity of indoxyl sulfate and p-cresyl sulfate: a systematic review.
J Am Soc Nephrol, 25 (2014), pp. 1897-1907
[52]
H.A. Mutsaers, E.G. Stribos, G. Glorieux, R. Vanholder, P. Olinga.
Chronic kidney disease and fibrosis: the role of uremic retention solutes.
Front Med (Lausanne), (2015),
[53]
B.K. Meijers, P. Evenepoel.
The gut-kidney axis: indoxyl sulfate, p-cresyl sulfate and CKD progression.
Nephrol Dial Transplant, 26 (2011), pp. 759-761
[54]
M. Motojima, A. Hosokawa, H. Yamato, T. Muraki, T. Yoshioka.
Uremic toxins of organic anions up-regulate PAI-1 expression by induction of NF-kappaB and free radical in proximal tubular cells.
Kidney Int, 63 (2003), pp. 1671-1680
[55]
I.W. Wu, K.H. Hsu, C.C. Lee, C.Y. Sun, H.J. Hsu, C.J. Tsai, et al.
p-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease.
Nephrol Dial Transplant, 26 (2011), pp. 938-947
[56]
J.R. Stubbs, J.A. House, A.J. Ocque, S. Zhang, C. Johnson, C. Kimber, et al.
Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden.
J Am Soc Nephrol, 27 (2016), pp. 305-313
Epub 2015 Jul 30
[57]
F.C. Barreto, D.V. Barreto, S. Liabeuf, N. Meert, G. Glorieux, M. Temmar, European Uremic Toxin Work Group (EUTox), et al.
Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients.
Clin J Am Soc Nephrol, 4 (2009), pp. 1551-1558
[58]
A.C. Raff, T.W. Meyer, T.H. Hostetter.
New insights into uremic toxicity.
Curr Opin Nephrol Hypertens, 17 (2008), pp. 560-565
[59]
V. Faure, L. Dou, F. Sabatier, C. Cerini, J. Sampol, Y. Berland, et al.
Elevation of circulating endothelial microparticles in patients with chronic renal failure.
J Thromb Haemost, 4 (2006), pp. 566-573
[60]
S. Lekawanvijit, A. Adrahtas, D.J. Kelly, A.R. Kompa, B.H. Wang, H. Krum.
Does indoxyl sulfate, a uraemic toxin, have direct effects on cardiac fibroblasts and myocytes?.
Eur Heart J, 31 (2010), pp. 1771-1779
[61]
K. Aoki, Y. Teshima, H. Kondo, S. Saito, A. Fukui, N. Fukunaga, et al.
Role of indoxyl sulfate as a predisposing factor for atrial fibrillation in renal dysfunction.
J Am Heart Assoc, 4 (2015), pp. e002023
[62]
P. Gross, Z.A. Massy, L. Henaut, C. Boudot, J. Cagnard, C. March, et al.
Para-cresyl sulfate acutely impairs vascular reactivity and induces vascular remodeling.
J Cell Physiol, 230 (2015), pp. 2927-2935
[63]
R. Poesen, L. Viaene, K. Verbeke, P. Augustijns, B. Bammens, K. Claes, et al.
Cardiovascular disease relates to intestinal uptake of p-cresol in patients with chronic kidney disease.
BMC Nephrol, 9 (2014), pp. 87
[64]
B.K. Meijers, B. Bammens, B. de Moor, K. Verbeke, Y. Vanrenterghem, P. Evenepoel.
Free p-cresol is associated with cardiovascular disease in hemodialysis patients.
Kidney Int, 73 (2008), pp. 1174-1180
[65]
C.J. Lin, C.F. Pan, H.L. Liu, C.K. Chuang, T. Jayakumar, T.J. Wang, et al.
The role of protein-bound uremic toxins on peripheral artery disease and vascular access failure in patients on hemodialysis.
Atherosclerosis, 225 (2012), pp. 173-179
[66]
C.J. Lin, V. Wu, P.C. Wu, C.J. Wu.
Meta-analysis of the associations of p-cresyl sulfate (PCS) and indoxyl sulfate (IS) with cardiovascular events and all-cause mortality in patients with chronic renal failure.
PLOS ONE, 10 (2015), pp. e0132589
[67]
L. Dou, M. Sallée, C. Cerini, S. Poitevin, B. Gondouin, N. Jourde-Chiche, et al.
The cardiovascular effect of the uremic solute indole-3 acetic acid.
J Am Soc Nephrol, 26 (2015), pp. 876-887
[68]
W.H. Tang, Z. Wang, D.J. Kennedy, Y. Wu, J.A. Buffa, B. Agatisa-Boyle, 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, 116 (2015), pp. 448-455
[69]
C. Missailidis, J. Hällqvist, A.R. Qureshi, P. Barany, O. Heimbürger, B. Lindholm, et al.
Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease.
PLOS ONE, 11 (2016), pp. e0141738
[70]
G.A. Kaysen, K.L. Johansen, G.M. Chertow, L.S. Dalrymple, J. Kornak, B. Grimes, et al.
Associations of trimethylamine N-oxide with nutritional and inflammatory biomarkers and cardiovascular outcomes in patients new to dialysis.
J Ren Nutr, 25 (2015), pp. 351-356
[71]
C.K. Chiang, T. Tanaka, R. Inagi, T. Fujita, M. Nangaku.
Indoxyl sulfate, a representative uremic toxin, suppresses erythropoietin production in a HIF-dependent manner.
Lab Invest, 91 (2011), pp. 1564-1571
[72]
M. Nangaku, I. Mimura, J. Yamaguchi, Y. Higashijima, T. Wada, T. Tanaka.
Role of uremic toxins in erythropoiesis-stimulating agent resistance in chronic kidney disease and dialysis patients.
J Ren Nutr, 25 (2015), pp. 160-163
[73]
M.S. Ahmed, M. Abed, J. Voelkl, F. Lang.
Triggering of suicidal erythrocyte death by uremic toxin indoxyl sulfate.
BMC Nephrol, 14 (2013), pp. 244
[74]
K. Yoshida, T. Yoneda, S. Kimura, K. Fujimoto, E. Okajima, Y. Hirao.
Polyamines as an inhibitor on erythropoiesis of hemodialysis patients by in vitro bioassay using the fetal mouse liver assay.
Ther Apher Dial, 10 (2006), pp. 267-272
[75]
T. Nii-Kono, Y. Iwasaki, M. Uchida, A. Fujieda, A. Hosokawa, M. Motojima, et al.
Indoxyl sulfate induces skeletal resistance to parathyroid hormone in cultured osteoblastic cells.
Kidney Int, 71 (2007), pp. 738-743
[76]
C.J. Lin, C.F. Pan, C.K. Chuang, H.L. Liu, F.J. Sun, T.J. Wang, et al.
Association of indoxyl sulfate with fibroblast growth factor 23 in patients with advanced chronic kidney disease.
Am J Med Sci, 347 (2014), pp. 370-376
[77]
J. Hirata, K. Hirai, H. Asai, C. Matsumoto, M. Inada, C. Miyaura, et al.
Indoxyl sulfate exacerbates low bone turnover induced by parathyroidectomy in young adult rats.
[78]
C.O. Soulage, L. Koppe, D. Fouque.
Protein-bound uremic toxins… new targets to prevent insulin resistance and dysmetabolism in patients with chronic kidney disease.
J Ren Nutr, 23 (2013), pp. 464-466
[79]
A. Ramezani, D.S. Raj.
The gut microbiome, kidney disease, and targeted interventions.
J Am Soc Nephrol, 25 (2014), pp. 657-670
[80]
M. Rossi, D.W. Johnson, H. Xu, J.J. Carrero, E. Pascoe, C. French, et al.
Dietary protein–fiber ratio associates with circulating levels of indoxyl sulfate and p-cresyl sulfate in chronic kidney disease patients.
Nutr Metab Cardiovasc Dis, 25 (2015), pp. 860-865
[81]
K.P. Patel, F.J. Luo, N.S. Plummer, T.H. Hostetter, T.W. Meyer.
The production of p-cresol sulfate and indoxyl sulfate in vegetarians versus omnivores.
Clin J Am Soc Nephrol, 7 (2012), pp. 982-988
[82]
S. Marzocco, F. Dal Piaz, L. Di Micco, S. Torraca, M.L. Sirico, D. Tartaglia, et al.
Very low protein diet reduces indoxyl sulfate levels in chronic kidney disease.
Blood Purif, 35 (2013), pp. 196-201
[83]
B.K. Meijers, V. de Preter, K. Verbeke, Y. Vanrenterghem, P. Evenepoel.
p-Cresyl sulfate serum concentrations in haemodialysis patients are reduced by the prebiotic oligofructose enriched inulin.
Nephrol Dial Transplant, 25 (2010), pp. 219-224
[84]
T.L. Sirich, N.S. Plummer, C.D. Gardner, T.H. Hostetter, T.W. Meyer.
Effect of increasing dietary fiber on plasma levels of colon-derived solutes in hemodialysis patients.
Clin J Am Soc Nephrol, 9 (2014), pp. 1603-1610
[85]
N.D. Vaziri, S.M. Liu, W.L. Lau, M. Khazaeli, S. Nazertehrani, S.H. Farzaneh, et al.
High amylose resistant starch diet ameliorates oxidative stress, inflammation, and progression of chronic kidney disease.
[86]
The effect of arabinoxylan-oligosaccharides (AXOS) on intestinal generation of microbial metabolites in chronic kidney disease. Available from: https://clinicaltrials.gov/ct2/show/study/NCT02141815?term5Bj%C3%B6rn1Meijers&rank54 2015 [accessed 20.03.16].
[87]
L. Koppe, D. Mafra, D. Fouque.
Probiotics and chronic kidney disease.
Kidney Int, 88 (2015), pp. 958-966
[88]
M. Hida, Y. Aiba, S. Sawamura, N. Suzuki, T. Satoh, Y. Koya.
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, 74 (1996), pp. 349-355
[89]
M.L. Simenhoff, S.R. Dunn, G.P. Zollner, M.E. Fitzpatrick, S.M. Emery, W.E. Sandine, 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, 22 (1996), pp. 92-96
[90]
F. Takayama, K. Taki, T. Niwa.
Bifidobacterium in gastro-resistant seamless capsule reduces serum levels of indoxyl sulfate in patients on hemodialysis.
Am J Kidney Dis, 41 (2003), pp. S142-S145
[91]
Y. Ando, Y. Miyata, K. Tanba, O. Saito, S. Muto, M. Kurosu, 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, 45 (2003), pp. 759-764
[92]
K. Taki, F. Takayama, T. Niwa.
Beneficial effects of Bifidobacteria in a gastroresistant seamless capsule on hyperhomocysteinemia in hemodialysis patients.
J Ren Nutr, 15 (2005), pp. 77-80
[93]
N. Ranganathan, E.A. Friedman, P. Tam, V. Rao, P. Ranganathan, R. Dheer.
Probiotic dietary supplementation in patients with stage 3 and 4 chronic kidney disease: a 6-month pilot scale trial in Canada.
Curr Med Res Opin, 25 (2009), pp. 1919-1930
[94]
N. Ranganathan, P. Ranganathan, E.A. Friedman, A. Joseph, B. Delano, D.S. Goldfarb, et al.
Pilot study of probiotic dietary supplementation for promoting healthy kidney function in patients with chronic kidney disease.
Adv Ther, 27 (2010), pp. 634-647
[95]
P.V. Miranda Alatriste, R. Urbina Arronte, C.O. Gómez Espinosa, M.Á. Espinosa Cuevas.
Effect of probiotics on human blood urea levels in patients with chronic renal failure.
Nutr Hosp, 29 (2014), pp. 582-590
[96]
I.-K. Wang, Y.Y. Wu, Y.F. Yang, I.W. Ting, C.C. Lin, T.H. Yen, 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, 6 (2015), pp. 423-430
[97]
R. Natarajan, B. Pechenyak, U. Vyas, P. Ranganathan, A. Weinberg, P. Liang, et al.
Randomized controlled trial of strain-specific probiotic fórmulation (Renadyl) in dialysis patients.
Biomed Res Int, 2014 (2014), pp. 568571
[98]
I. Nakabayashi, M. Nakamura, K. Kawakami, T. Ohta, I. Kato, K. Uchida, et al.
Effects of synbiotic treatment on serum level of p-cresol in haemodialysis patients: a preliminary study.
Nephrol Dial Transplant, 26 (2010), pp. 1094-1098
[99]
T. Ogawa, M. Shimada, N. Nagano, K. Ito, T. Ando, Y. Shimomura, et al.
Oral administration of Bifidobacterium longum in a gastro-resistant seamless capsule decreases serum phosphate levels in patients receiving haemodialysis.
Clin Kidney J, 5 (2012), pp. 373-374
[100]
M. Pavan.
Influence of prebiotic and probiotic supplementation on the progression of chronic kidney disease.
Minerva Urol Nephrol, (2014),
[101]
J. Cruz-Mora, N.E. Martínez-Hernández, F. Martín del Campo-López, D. Viramontes-Hörner, B. Vizmanos-Lamotte, J.F. Muñoz-Valle, et al.
Effects of a symbiotic on gut microbiota in Mexican patients with end-stage renal disease.
J Ren Nutr, 24 (2014), pp. 330-335
[102]
B. Guida, R. Germanò, R. Trio, D. Russo, B. Memoli, L. Grumetto.
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, 24 (2014), pp. 1043-1049
[103]
D. Viramontes-Hörner, F. Márquez-Sandoval, F. Martín-del-Campo, B. Vizmanos-Lamotte, A. Sandoval-Rodríguez, J. Armendáriz-Borunda, et al.
Effect of a symbiotic gel (Lactobacillus acidophilus+Bifidobacterium lactis+Inulin) on presence and severity of gastrointestinal symptoms in hemodialysis patients.
J Ren Nutr, 25 (2015), pp. 284-291
[104]
M. Rossi, D.W. Johnson, M. Morrison, E.M. Pascoe, J.S. Coombes, J.M. Forbes, et al.
Synbiotics Easing Renal Failure by Improving Gut Microbiology (SYNERGY): a randomised cross-over trial.
Clin J Am Soc Nephrol, 11 (2016), pp. 223-231
[105]
G. Schulman, R. Agarwal, M. Acharya, T. Berl, S. Blumenthal, N. Kopyt.
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, 47 (2006), pp. 565-577
[106]
S. Yamamoto, J.J. Kazama, K. Omori, K. Matsuo, Y. Takahashi, K. Kawamura, et al.
Continuous reduction of protein-bound uraemic toxins with improved oxidative stress by using the oral charcoal adsorbent AST-120 in haemodialysis patients.
Sci Rep, 5 (2015), pp. 14381
[107]
I.W. Wu, K.H. Hsu, C.Y. Sun, C.J. Tsai, M.S. Wu, C.C. Lee.
Oral adsorbent AST-120 potentiates the effect of erythropoietin-stimulating agents on stage 5 chronic kidney disease patients: a randomized crossover study.
Nephrol Dial Transplant, 29 (2014), pp. 1719-1727
[108]
N.D. Vaziri, J. Yuan, M. Khazaeli, Y. Masuda, H. Ichii, S. Liu.
Oral activated charcoal adsorbent (AST-120) ameliorates chronic kidney disease-induced intestinal epithelial barrier disruption.
Am J Nephrol, 37 (2013), pp. 518-525
[109]
G. Schulman, T. Berl, G.J. Beck, G. Remuzzi, E. Ritz, K. Arita, et al.
Randomized placebo controlled EPPIC trials of AST-120 in CKD.
J Am Soc Nephrol, 26 (2015), pp. 1732-1746
[110]
E. Sato, A. Tanaka, J.I. Oyama, A. Yamasaki, M. Shimomura, A. Hiwatashi, et al.
Long-term effects of AST-120 on the progression and prognosis of pre-dialysis chronic kidney disease: a 5-year retrospective study.
Heart Vessels, 31 (2016), pp. 1625-1632
[111]
R. De Smet, F. Thermote, N. Lameire.
Sevelamer hydrochloride adsorbs the uremic compound indoxyl sulfate.
J Am Soc Nephrol, (2003), pp. 208
ASN Renal Week. Abstract F-PO-660
[112]
A.E. Stinghen, S.M. Gonçalves, S. Bucharles, F.S. Branco, B. Gruber, A.B. Hauser, et al.
Sevelamer decreases systemic inflammation in parallel to a reduction in endotoxemia.
Blood Purif, 29 (2010), pp. 352-356
[113]
J.F. Navarro-González, C. Mora-Fernández, M. Muros de Fuentes, J. Donate-Correa, V. Cazaña-Pérez, J. García-Pérez.
Effect of phosphate binders on serum inflammatory profile, soluble CD14, and endotoxin levels in hemodialysis patients.
Clin J Am Soc Nephrol, 6 (2011), pp. 2272-2279

Please cite this article as: Guldris SC, Parra EG, Amenós AC. Microbiota intestinal en la enfermedad renal crónica. Nefrología. 2017;37:9–19.

Copyright © 2016. Sociedad Española de Nefrología
Idiomas
Nefrología (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?