Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Curr Nutr Rep ; 11(4): 765-779, 2022 12.
Article in English | MEDLINE | ID: mdl-36138326

ABSTRACT

PURPOSE OF REVIEW: This narrative review aimed to summarize the current evidence on the connection between dysbiosis and vitamin K deficiency in patients with chronic kidney disease (CKD). The presence of dysbiosis (perturbations in the composition of the microbiota) has been described in several non-communicable diseases, including chronic kidney disease, and it has been hypothesized that dysbiosis may cause vitamin K deficiency. Patients with CKD present both vitamin K deficiency and gut dysbiosis; however, the relationship between gut dysbiosis and vitamin K deficiency remains to be addressed. RECENT FINDINGS: Recently, few studies in animals have demonstrated that a dysbiotic environment is associated with low production of vitamin K by the gut microbiota. Vitamin K plays a vital role in blood coagulation as well as in the cardiovascular and bone systems. It serves as a cofactor for γ-glutamyl carboxylases and thus is essential for the post-translational modification and activation of vitamin K-dependent calcification regulators, such as osteocalcin, matrix Gla protein, Gla-rich protein, and proteins C and S. Additionally, vitamin K executes essential antioxidant and anti-inflammatory functions. Dietary intake is the main source of vitamin K; however, it also can be produced by gut microbiota. This review discusses the effects of uremia on the imbalance in gut microbiota, vitamin K-producing bacteria, and vitamin K deficiency in CKD patients, leading to a better understanding and raising hypothesis for future clinical studies.


Subject(s)
Renal Insufficiency, Chronic , Uremia , Vitamin K Deficiency , Animals , Humans , Dysbiosis , Vitamin K/metabolism , Renal Insufficiency, Chronic/microbiology , Uremia/metabolism , Uremia/microbiology , Vitamin K Deficiency/complications , Vitamin K Deficiency/metabolism
2.
Microbiol Spectr ; 10(1): e0194221, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35171026

ABSTRACT

Lung transplant recipients (LTRs) are vulnerable to hyperammonemia syndrome (HS) in the early postoperative period, a condition typically unresponsive to nonantibiotic interventions. HS in LTRs is strongly correlated with Ureaplasma infection of the respiratory tract, although it is not well understood what makes LTRs preferentially susceptible to HS compared to other immunocompromised hosts. Ureaplasma species harbor highly active ureases, and postoperative LTRs commonly experience uremia. We hypothesized that uremia could be a potentiating comorbidity, providing increased substrate for ureaplasmal ureases. Using a novel dialyzed flow system, the ammonia-producing capacities of four isolates of Ureaplasma parvum and six isolates of Ureaplasma urealyticum in media formulations relating to normal and uremic host conditions were tested. For all isolates, growth under simulated uremic conditions resulted in increased ammonia production over 24 h, despite similar endpoint bacterial quantities. Further, transcripts of ureC (from the ureaplasmal urease gene cluster) from U. urealyticum IDRL-10763 and ATCC-27816 rose at similar rates under uremic and nonuremic conditions, with similar endpoint populations under the two conditions (despite markedly increased ammonia concentrations under uremic conditions), indicating that the difference in ammonia production by these isolates is due to increased urease activity, not expression. Lastly, uremic mice infected with an Escherichia coli strain harboring a U. urealyticum serovar 8 gene cluster exhibited higher blood ammonia levels compared to nonuremic mice infected with the same strain. Taken together, these data show that U. urealyticum and U. parvum produce more ammonia under uremic conditions compared to nonuremic conditions. This implies that uremia is a plausible contributing factor to Ureaplasma-induced HS in LTRs. IMPORTANCE Ureaplasma-induced hyperammonemia syndrome is a deadly complication affecting around 4% of lung transplant recipients and, to a lesser extent, other solid organ transplant patients. Understanding the underlying mechanisms will inform patient management, potentially decreasing mortality and morbidity. Here, it is shown that uremia is a plausible contributing factor to the pathophysiology of the condition.


Subject(s)
Hyperammonemia/complications , Hyperammonemia/microbiology , Uremia/complications , Uremia/microbiology , Ammonia/metabolism , Animals , Humans , Immunocompromised Host , Lung , Lung Transplantation , Mice , Transplant Recipients , Ureaplasma , Ureaplasma urealyticum/isolation & purification , Urinary Tract
3.
Toxins (Basel) ; 13(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807343

ABSTRACT

Numerous studies have indicated that the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD) is strictly associated with the accumulation of toxic metabolites in blood and other metabolic compartments. This accumulation was suggested to be related to enhanced generation of toxins from the dysbiotic microbiome accompanied by their reduced elimination by impaired kidneys. Intestinal microbiota play a key role in the accumulation of uremic toxins due to the fact that numerous uremic solutes are generated in the process of protein fermentation by colonic microbiota. Some disease states, including CKD, are associated with the presence of dysbiosis, which can be defined as an "imbalanced intestinal microbial community with quantitative and qualitative changes in the composition and metabolic activities of the gut microbiota". The results of studies have confirmed the altered composition and functions of gut microbial community in chronic kidney disease. In the course of CKD protein-bound uremic toxins, including indoxyl sulfate, p-cresyl glucuronide, p-cresyl sulfate and indole-3-acetic acid are progressively accumulated. The presence of chronic kidney disease may be accompanied by the development of intestinal inflammation and epithelial barrier impairment leading to hastened systemic translocation of bacterial-derived uremic toxins and consequent oxidative stress injury to the kidney, cardiovascular and endocrine systems. These findings offer new therapeutic possibilities for the management of uremia, inflammation and kidney disease progression and the prevention of adverse outcomes in CKD patients. It seems that dietary interventions comprising prebiotics, probiotics, and synbiotics could pose a promising strategy in the management of uremic toxins in CKD.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Renal Insufficiency, Chronic/blood , Toxins, Biological/blood , Uremia/blood , Animals , Dietary Supplements , Disease Progression , Dysbiosis , Host-Pathogen Interactions , Humans , Renal Dialysis , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/therapy , Uremia/diagnosis , Uremia/microbiology , Uremia/therapy
4.
Toxins (Basel) ; 13(3)2021 03 19.
Article in English | MEDLINE | ID: mdl-33808581

ABSTRACT

Imbalanced colonic microbial metabolism plays a pivotal role in generating protein-bound uraemic toxins (PBUTs), which accumulate with deteriorating kidney function and contribute to the uraemic burden of children with chronic kidney disease (CKD). Dietary choices impact the gut microbiome and metabolism. The aim of this study was to investigate the relation between dietary fibre and gut-derived PBUTs in paediatric CKD. Sixty-one (44 male) CKD children (9 ± 5 years) were prospectively followed for two years. Dietary fibre intake was evaluated by either 24-h recalls (73%) or 3-day food records (27%) at the same time of blood sampling for assessment of total and free serum levels of different PBUTs using liquid chromatography. We used linear mixed models to assess associations between fibre intake and PBUT levels. We found an inverse association between increase in fibre consumption (g/day) and serum concentrations of free indoxyl sulfate (-3.1% (-5.9%; -0.3%) (p = 0.035)), free p-cresyl sulfate (-2.5% (-4.7%; -0.3%) (p = 0.034)), total indole acetic acid (IAA) (-1.6% (-3.0%; -0.3%) (p = 0.020)), free IAA (-6.6% (-9.3%; -3.7%) (p < 0.001)), total serum p-cresyl glucuronide (pCG) (-3.0% (-5.6%; -0.5%) (p = 0.021)) and free pCG levels (-3.3% (-5.8%; -0.8%) (p = 0.010)). The observed associations between dietary fibre intake and the investigated PBUTs highlight potential benefits of fibre intake for the paediatric CKD population. The present observational findings should inform and guide adaptations of dietary prescriptions in children with CKD.


Subject(s)
Bacteria/metabolism , Dietary Fiber/administration & dosage , Gastrointestinal Microbiome , Intestines/microbiology , Renal Insufficiency, Chronic/diet therapy , Toxins, Biological/blood , Uremia/diet therapy , Adolescent , Age Factors , Belgium , Child , Child, Preschool , Dysbiosis , Female , Humans , Infant , Longitudinal Studies , Male , Prognosis , Prospective Studies , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/microbiology , Uremia/blood , Uremia/diagnosis , Uremia/microbiology
5.
Toxins (Basel) ; 13(3)2021 03 18.
Article in English | MEDLINE | ID: mdl-33803899

ABSTRACT

Patients with chronic kidney disease (CKD) are at an increased risk of thromboembolic complications, including myocardial infarction, stroke, deep vein thrombosis, and pulmonary embolism. These complications lead to increased mortality. Evidence points to the key role of CKD-associated dysbiosis and its effect via the generation of gut microbial metabolites in inducing the prothrombotic phenotype. This phenomenon is known as thrombolome, a panel of intestinal bacteria-derived uremic toxins that enhance thrombosis via increased tissue factor expression, platelet hyperactivity, microparticles release, and endothelial dysfunction. This review discusses the role of uremic toxins derived from gut-microbiota metabolism of dietary tryptophan (indoxyl sulfate (IS), indole-3-acetic acid (IAA), kynurenine (KYN)), phenylalanine/tyrosine (p-cresol sulfate (PCS), p-cresol glucuronide (PCG), phenylacetylglutamine (PAGln)) and choline/phosphatidylcholine (trimethylamine N-oxide (TMAO)) in spontaneously induced thrombosis. The increase in the generation of gut microbial uremic toxins, the activation of aryl hydrocarbon (AhRs) and platelet adrenergic (ARs) receptors, and the nuclear factor kappa B (NF-κB) signaling pathway can serve as potential targets during the prevention of thromboembolic events. They can also help create a new therapeutic approach in the CKD population.


Subject(s)
Bacteria/metabolism , Blood Coagulation , Gastrointestinal Microbiome , Intestines/microbiology , Renal Insufficiency, Chronic/complications , Thromboembolism/etiology , Toxins, Biological/blood , Uremia/complications , Animals , Dysbiosis , Humans , Prognosis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/microbiology , Risk Factors , Thromboembolism/blood , Thromboembolism/microbiology , Uremia/blood , Uremia/microbiology
6.
Toxins (Basel) ; 13(4)2021 04 10.
Article in English | MEDLINE | ID: mdl-33920096

ABSTRACT

Chronic kidney disease (CKD) affects more than 10% of the world population and leads to excess morbidity and mortality (with cardiovascular disease as a leading cause of death). Vascular calcification (VC) is a phenomenon of disseminated deposition of mineral content within the media layer of arteries preceded by phenotypic changes in vascular smooth muscle cells (VSMC) and/or accumulation of mineral content within the atherosclerotic lesions. Medial VC results in vascular stiffness and significantly contributes to increased cardio-vascular (CV) morbidity, whereas VC of plaques may rather increase their stability. Mineral and bone disorders of CKD (CKD-MBD) contribute to VC, which is further aggravated by accumulation of uremic toxins. Both CKD-MBD and uremic toxin accumulation affect not only patients with advanced CKD (glomerular filtration rate (GFR) less than 15 mL/min./1.72 m2, end-stage kidney disease) but also those on earlier stages of a disease. The key uremic toxins that contribute to VC, i.e., p-cresyl sulphate (PCS), indoxyl sulphate (IS) and trimethylamine-N-oxide (TMAO) originate from bacterial metabolism of gut microbiota. All mentioned toxins promote VC by several mechanisms, including: Transdifferentiation and apoptosis of VSMC, dysfunction of endothelial cells, oxidative stress, interaction with local renin-angiotensin-aldosterone system or miRNA profile modification. Several attractive methods of gut microbiota manipulations have been proposed in order to modify their metabolism and to limit vascular damage (and VC) triggered by uremic toxins. Unfortunately, to date no such method was demonstrated to be effective at the level of "hard" patient-oriented or even clinically relevant surrogate endpoints.


Subject(s)
Bacteria/metabolism , Blood Vessels/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Vascular Calcification/metabolism , Animals , Blood Vessels/pathology , Chronic Kidney Disease-Mineral and Bone Disorder/microbiology , Disease Progression , Humans , Prognosis , Renal Insufficiency, Chronic/microbiology , Uremia/microbiology , Vascular Calcification/microbiology , Vascular Calcification/pathology
7.
Immunohorizons ; 5(1): 16-24, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33451988

ABSTRACT

Infections are the second major cause of mortality in patients with kidney disease and accompanying uremia. Both vascular access and non-access-related infections contribute equally to the infection-related deaths in patients with kidney disease. Dialysis is the most common cause of systemic infection by Candida albicans in these patients. C albicans also reside in the gastrointestinal tract as a commensal fungus. However, the contribution of gut-derived C albicans in non-access-related infections in kidney disease is unknown. Using a mouse model of kidney disease, we demonstrate that uremic animals showed increased gut barrier permeability, impaired mucosal defense, and dysbiosis. The disturbance in gut homeostasis is sufficient to drive the translocation of microbiota and intestinal pathogen Citrobacter rodentium to extraintestinal sites but not C albicans Interestingly, a majority of uremic animals showed fungal translocation only when the gut barrier integrity is disrupted. Our data demonstrate that uremia coupled with gut mucosal damage may aid in the translocation of C. albicans and cause systemic infection in kidney disease. Because most of the individuals with kidney disease suffer from some form of gut mucosal damage, these results have important implications in the risk stratification and control of non-access-related opportunistic fungal infections in these patients.


Subject(s)
Candida albicans/physiology , Citrobacter rodentium/physiology , Intestinal Mucosa/microbiology , Intestines/microbiology , Uremia/microbiology , Animals , Citrobacter rodentium/growth & development , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions , Humans , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Symbiosis
8.
Toxins (Basel) ; 12(12)2020 12 11.
Article in English | MEDLINE | ID: mdl-33322362

ABSTRACT

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


Subject(s)
Acute Kidney Injury/chemically induced , Gastrointestinal Microbiome/drug effects , Toxins, Biological/toxicity , Uremia/chemically induced , Acute Kidney Injury/microbiology , Acute Kidney Injury/physiopathology , Animals , Gastrointestinal Microbiome/physiology , Humans , Microbiota/drug effects , Microbiota/physiology , Uremia/microbiology , Uremia/physiopathology
9.
Toxins (Basel) ; 12(9)2020 09 11.
Article in English | MEDLINE | ID: mdl-32932981

ABSTRACT

Chronic kidney disease (CKD) afflicts more than 500 million people worldwide and is one of the fastest growing global causes of mortality. When glomerular filtration rate begins to fall, uremic toxins accumulate in the serum and significantly increase the risk of death from cardiovascular disease and other causes. Several of the most harmful uremic toxins are produced by the gut microbiota. Furthermore, many such toxins are protein-bound and are therefore recalcitrant to removal by dialysis. We review the derivation and pathological mechanisms of gut-derived, protein-bound uremic toxins (PBUTs). We further outline the emerging relationship between kidney disease and gut dysbiosis, including the bacterial taxa altered, the regulation of microbial uremic toxin-producing genes, and their downstream physiological and neurological consequences. Finally, we discuss gut-targeted therapeutic strategies employed to reduce PBUTs. We conclude that targeting the gut microbiota is a promising approach for the treatment of CKD by blocking the serum accumulation of PBUTs that cannot be eliminated by dialysis.


Subject(s)
Bacteria/metabolism , Bacterial Toxins/metabolism , Gastrointestinal Microbiome , Renal Insufficiency, Chronic/microbiology , Uremia/microbiology , Animals , Disease Progression , Dysbiosis , Humans , Protein Binding , Renal Dialysis , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Uremia/metabolism , Uremia/therapy
10.
Biomed Res Int ; 2020: 6739762, 2020.
Article in English | MEDLINE | ID: mdl-32685517

ABSTRACT

BACKGROUND: To investigate the effect of hemodialysis on microbiota in uremic patients. OBJECTIVE: To investigate the effect of hemodialysis on microbiota in uremic patients. METHODS: This study included 85 adult patients who have received hemodialysis since August 2014, and the treatment plan has not changed for more than 12 months. These patients were divided into hemodialysis group (group A), hemodialysis+hemodialysis filtration group (group B), and hemodialysis+hemodialysis filtration+blood perfusion group (group C). Twenty-four adult ESRD patients (CK group) were enrolled. Serum biochemical indexes were measured, glomerular filtration rate (EGFR) was estimated, dialysis adequacy (kt/V) was calculated, and fresh feces were collected. At the same time, the feces of 30 health workers were selected as the control. 16S rRNA sequence was used to determine the intestinal flora of all fecal specimens. First of all, we analyzed the difference of the whole flora distribution between dialysis and nondialysis ESRD patients; then, we selected the most representative content of bifidobacteria, Lactobacillus acidophilus, Escherichia coli, and Enterococcus faecalis to analyze the influence of different blood purification methods on the intestinal flora. RESULTS: (1) The level of C-reactive protein (CRP) in dialysis patients was lower than that in nondialysis ESRD patients, and CRP in group C was lower than that in groups A and B. There was no significant difference in kt/V between group A, group B, and group C. There was no significant difference in EGFR between the four groups. (2) The species diversity of ESRD patients without dialysis (CK group) was significantly lower than that of ESRD patients with dialysis; there was no significant difference between group A and group B; the species diversity of group C was significantly higher than that of group A and group B. (3) Compared with the control group, the levels of bifidobacteria and Lactobacillus acidophilus in ESRD patients were significantly lower, while the levels of Escherichia coli and Enterococcus faecalis were significantly higher. (4) The levels of bifidobacteria and Lactobacillus acidophilus in hemodialysis patients were significantly higher than those in nonblood purification treatment group, and the levels of Escherichia coli and Enterococcus faecalis were significantly lower than those in nonblood purification treatment group. (5) The level of Lactobacillus acidophilus in group C was significantly higher than that in groups A and B, and the level of Escherichia coli was significantly lower than that in groups A and B. CONCLUSION: ESRD patients have microbiota disorder. Hemodialysis can improve microbiota disorder in uremic patients. Compared with ordinary hemodialysis, combined hemoperfusion dialysis can further improve microbiota disorder.


Subject(s)
Gastrointestinal Microbiome , Kidney Failure, Chronic/microbiology , Kidney Failure, Chronic/therapy , Renal Dialysis/methods , Uremia/microbiology , Uremia/therapy , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Feces/microbiology , Female , Humans , Kidney Failure, Chronic/blood , Male , Middle Aged , RNA, Ribosomal, 16S/genetics , Uremia/pathology
11.
Int J Pharm ; 584: 119450, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32464229

ABSTRACT

The progressive loss of renal function in chronic kidney disease (CKD) leads to the accumulation of uremic toxins. Recent studies related uremic plasma as well dysbiotic gut microbiome to impaired intestinal barrier function, allowing the translocation of microorganisms or by-products from the intestinal lumen to systemic circulation, contributing to systemic inflammation, cardiovascular risk and progression of CKD. Our main goal was to evaluate the impact of different uremic conditions on an improved in vitro intestinal Caco-2/HT29-MTX/Raji B triple co-culture model. For that, the impact of CKD patients' plasma and elevated urea concentration and its by-products on the triple model was assessed. The results showed that uremic conditions did not potentiate the Escherichia coli (E. coli) translocation, although may interfere with the integrity and the permeability of the intestinal barrier. Also, results showed that E. coli translocation was higher in Caco-2 monoculture than in Caco-2/HT29-MTX/Raji B triple model, suggesting that the triple model creates a more effective intestinal barrier. This study allowed to conclude that the uremic state influences the integrity of the intestinal barrier, but this influence could not be directly translated in an increase on the E. coli translocation through the intestinal epithelium, at least in Caco-2/HT29-MTX/Raji B intestinal epithelial barrier model.


Subject(s)
Bacterial Translocation , Escherichia coli/physiology , Intestinal Mucosa/microbiology , Uremia/microbiology , Cell Line, Tumor , Coculture Techniques , Humans , Intestinal Mucosa/metabolism , Permeability , Tight Junctions , Uremia/metabolism
12.
Toxins (Basel) ; 12(6)2020 05 27.
Article in English | MEDLINE | ID: mdl-32471179

ABSTRACT

Gut microbial metabolism is not only an important source of uremic toxins but may also help to maintain the vitamin K stores of the host. We hypothesized that sevelamer therapy, a commonly used phosphate binder in patients with end-stage kidney disease (ESKD), associates with a disturbed gut microbial metabolism. Important representatives of gut-derived uremic toxins, including indoxyl sulfate (IndS), p-Cresyl sulfate (pCS), trimethylamine N-oxide (TMAO), phenylacetylglutamine (PAG) and non-phosphorylated, uncarboxylated matrix-Gla protein (dp-ucMGP; a marker of vitamin K status), were analyzed in blood samples from 423 patients (65% males, median age 54 years) with ESKD. Demographics and laboratory data were extracted from electronic files. Sevelamer users (n = 172, 41%) were characterized by higher phosphate, IndS, TMAO, PAG and dp-ucMGP levels compared to non-users. Sevelamer was significantly associated with increased IndS, PAG and dp-ucMGP levels, independent of age, sex, calcium-containing phosphate binder, cohort, phosphate, creatinine and dialysis vintage. High dp-ucMGP levels, reflecting vitamin K deficiency, were independently and positively associated with PAG and TMAO levels. Sevelamer therapy associates with an unfavorable gut microbial metabolism pattern. Although the observational design precludes causal inference, present findings implicate a disturbed microbial metabolism and vitamin K deficiency as potential trade-offs of sevelamer therapy.


Subject(s)
Bacteria/drug effects , Bacterial Toxins/blood , Chelating Agents/adverse effects , Colon/microbiology , Gastrointestinal Microbiome/drug effects , Kidney Failure, Chronic/drug therapy , Sevelamer/adverse effects , Uremia/drug therapy , Vitamin K Deficiency/chemically induced , Adult , Aged , Bacteria/metabolism , Biomarkers/blood , Female , Humans , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/microbiology , Male , Middle Aged , Phosphates/blood , Risk Factors , Treatment Outcome , Uremia/blood , Uremia/microbiology , Vitamin K Deficiency/blood
13.
Toxins (Basel) ; 12(4)2020 04 11.
Article in English | MEDLINE | ID: mdl-32290429

ABSTRACT

Several of the uremic toxins, which are difficult to remove by dialysis, originate from the gut bacterial metabolism. This opens opportunities for novel targets trying to decrease circulating levels of these toxins and their pathophysiological effects. The current review focuses on immunomodulatory effects of these toxins both at their side of origin and in the circulation. In the gut end products of the bacterial metabolism such as p-cresol, trimethylamine and H2S affect the intestinal barrier structure and function while in the circulation the related uremic toxins stimulate cells of the immune system. Both conditions contribute to the pro-inflammatory status of patients with chronic kidney disease (CKD). Generation and/or absorption of these toxin precursors could be targeted to decrease plasma levels of their respective uremic toxins and to reduce micro-inflammation in CKD.


Subject(s)
Bacteria/metabolism , Bacterial Toxins/metabolism , Colon/microbiology , Gastrointestinal Microbiome , Renal Insufficiency, Chronic/microbiology , Uremia/microbiology , Animals , Bacterial Toxins/blood , Bacterial Toxins/immunology , Colon/immunology , Colon/metabolism , Dysbiosis , Host-Pathogen Interactions , Humans , Inflammation Mediators/blood , Intestinal Absorption , Permeability , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/immunology , Uremia/blood , Uremia/immunology
14.
Clin Exp Nephrol ; 23(7): 908-919, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30895529

ABSTRACT

BACKGROUND: Constipation is frequently observed in patients with chronic kidney disease (CKD). Lactulose is expected to improve the intestinal environment by stimulating bowel movements as a disaccharide laxative and prebiotic. We studied the effect of lactulose on renal function in adenine-induced CKD rats and monitored uremic toxins and gut microbiota. METHODS: Wistar/ST male rats (10-week-old) were fed 0.75% adenine-containing diet for 3 weeks to induce CKD. Then, they were divided into three groups and fed as follows: control, normal diet; and 3.0- and 7.5-Lac, 3.0% and 7.5% lactulose-containing diets, respectively, for 4 weeks. Normal diet group was fed normal diet for 7 weeks. The rats were observed for parameters including renal function, uremic toxins, and gut microbiota. RESULTS: The control group showed significantly higher serum creatinine (sCr) and blood urea nitrogen (BUN) 3 weeks after adenine feeding than at baseline, with a 8.5-fold increase in serum indoxyl sulfate (IS). After switching to 4 weeks of normal diet following adenine feeding, the sCr and BUN in control group remained high with a further increase in serum IS. In addition, tubulointerstitial fibrosis area was increased in control group. On the other hand, 3.0- and 7.5-Lac groups improved sCr and BUN levels, and suppressed tubulointerstitial fibrosis, suggesting preventing of CKD progression by lactulose. Lac groups also lowered level of serum IS and proportions of gut microbiota producing IS precursor. CONCLUSION: Lactulose modifies gut microbiota and ameliorates CKD progression by suppressing uremic toxin production.


Subject(s)
Adenine , Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Kidney/drug effects , Lactulose/pharmacology , Prebiotics , Renal Insufficiency, Chronic/prevention & control , Uremia/prevention & control , Animals , Bacteria/metabolism , Biomarkers/blood , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Disease Progression , Fibrosis , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Oxidative Stress/drug effects , Rats, Wistar , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/physiopathology , Uremia/chemically induced , Uremia/microbiology , Uremia/physiopathology
15.
Am J Physiol Renal Physiol ; 316(6): F1211-F1217, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30864840

ABSTRACT

The gut microbiome is composed of a diverse population of bacteria that have beneficial and adverse effects on human health. The microbiome has recently gained attention and is increasingly noted to play a significant role in health and a number of disease states. Increasing urea concentration during chronic kidney disease (CKD) leads to alterations in the intestinal flora that can increase production of gut-derived toxins and alter the intestinal epithelial barrier. These changes can lead to an acceleration of the process of kidney injury. A number of strategies have been proposed to interrupt this pathway of injury in CKD. The purpose of this review is to summarize the role of the gut microbiome in CKD, tools used to study this microbial population, and attempts to alter its composition for therapeutic purposes.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Kidney/metabolism , Renal Insufficiency, Chronic/microbiology , Urea/metabolism , Uremia/microbiology , Animals , Dietary Supplements , Host-Pathogen Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/physiopathology , Kidney/physiopathology , Permeability , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Uremia/metabolism , Uremia/physiopathology , Uremia/therapy
16.
Nat Rev Nephrol ; 15(5): 301-316, 2019 05.
Article in English | MEDLINE | ID: mdl-30728454

ABSTRACT

Uraemic syndrome (also known as uremic syndrome) in patients with advanced chronic kidney disease involves the accumulation in plasma of small-molecule uraemic solutes and uraemic toxins (also known as uremic toxins), dysfunction of multiple organs and dysbiosis of the gut microbiota. As such, uraemic syndrome can be viewed as a disease of perturbed inter-organ and inter-organism (host-microbiota) communication. Multiple biological pathways are affected, including those controlled by solute carrier (SLC) and ATP-binding cassette (ABC) transporters and drug-metabolizing enzymes, many of which are also involved in drug absorption, distribution, metabolism and elimination (ADME). The remote sensing and signalling hypothesis identifies SLC and ABC transporter-mediated communication between organs and/or between the host and gut microbiota as key to the homeostasis of metabolites, antioxidants, signalling molecules, microbiota-derived products and dietary components in body tissues and fluid compartments. Thus, this hypothesis provides a useful perspective on the pathobiology of uraemic syndrome. Pathways considered central to drug ADME might be particularly important for the body's attempts to restore homeostasis, including the correction of disturbances due to kidney injury and the accumulation of uraemic solutes and toxins. This Review discusses how the remote sensing and signalling hypothesis helps to provide a systems-level understanding of aspects of uraemia that could lead to novel approaches to its treatment.


Subject(s)
Renal Insufficiency, Chronic/physiopathology , Signal Transduction , Uremia/etiology , Biomarkers/metabolism , Host Microbial Interactions/physiology , Humans , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/microbiology , Uremia/immunology , Uremia/microbiology , Uremia/physiopathology
17.
J Nephrol ; 32(5): 733-740, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30673975

ABSTRACT

The gut microbiota is considered to be a novel important factor to take into account in the pathogenesis of chronic kidney disease and uremia. Much attention has been paid to specific uremic retention solutes of microbial origin, such as indoxyl sulfate, p-cresyl sulfate, and trimethylamine-N-oxide. However, other novel less well studied compounds, such as hydrogen sulfide and related sulfur metabolites (sulfane sulfur, lanthionine, etc.), should be included in a more comprehensive appraisal of this topic, in light of the potential therapeutic opportunities for the future.


Subject(s)
Gastrointestinal Microbiome/physiology , Sulfur Compounds/metabolism , Uremia/metabolism , Uremia/microbiology , Folic Acid/metabolism , Humans
18.
Semin Nephrol ; 39(1): 107-116, 2019 01.
Article in English | MEDLINE | ID: mdl-30606403

ABSTRACT

The recent surge in research on the intestinal microbiota has greatly changed our understanding of human biology. Significant technical advances in DNA sequencing analysis and its application to metagenomics and metatranscriptomics has profoundly enhanced our ability to quantify and track complex microbial communities and to begin understanding their impact on human health and disease. This has led to a better understanding of the relationships between the intestinal microbiome and renal physiology/pathophysiology. In this review, we discuss the interactions between intestinal microbiota and kidney. We focus on select aspects including the intestinal barrier, immunologic and soluble mediators of microbiome effects, and effects of dysbiosis on acute kidney injury. Relevant studies on microbiome changes in other renal diseases are highlighted. We also introduce potential mechanisms of intervention with regard to gut microbiota in renal diseases.


Subject(s)
Acute Kidney Injury/microbiology , Dysbiosis/complications , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/immunology , Acute Kidney Injury/immunology , Animals , Blood Pressure , Fatty Acids, Volatile/metabolism , Humans , Methylamines/metabolism , Prebiotics , Probiotics/therapeutic use , Uremia/microbiology
19.
Nephrol Dial Transplant ; 34(11): 1876-1884, 2019 11 01.
Article in English | MEDLINE | ID: mdl-29939302

ABSTRACT

BACKGROUND: Microbial-derived uremic toxins, p-cresyl sulfate (PCS), indoxyl sulfate (IS) and indole 3-acetic acid (IAA), have been associated with the burden of chronic kidney disease (CKD). Prebiotics have emerged as an alternative to modulate the gut environment and to attenuate toxin production. This trial aims to investigate the effect of a prebiotic fructooligosaccharide (FOS) on uremic toxins of non-dialysis-dependent CKD (NDD-CKD) patients. METHODS: A double-blind, placebo-controlled, randomized trial was conducted for 3 months. In all, 50 nondiabetic NDD-CKD patients [estimated glomerular filtration rate (eGFR) <45 mL/min/1.73 m2], aged 18-80 years, were allocated to prebiotic (FOS, 12 g/day) or placebo (maltodextrin, 12 g/day) groups. Primary outcomes were changes in serum (total and free) and urinary (total) PCS. Secondary outcomes included changes in IS, IAA, serum markers of intestinal permeability (zonulin), gut-trophic factors (epidermal growth factor and glucagon-like peptide-2), eGFR, inflammation (high sensitive c-reactive protein and interleukin-6), homeostatic model assessment-insulin resistance, lipid profile and gastrointestinal symptoms. RESULTS: From 50 participants (54% men, 57.3 ± 14.6 years and eGFR 21.4 ± 7.6 mL/min/1.73 m2), 46 completed the follow-up. No changes in dietary intake or gastrointestinal symptoms were observed. There was a trend in the difference of serum total ΔPCS (treatment effect adjusted for baseline levels: -12.4 mg/L; 95% confidence interval (-5.6 to 0.9 mg/L; P = 0.07) and serum-free Δ%PCS [intervention -8.6 (-41.5 to 13.9%) versus placebo 3.5 (-28.8 to 85.5%); P = 0.07] between the groups. The trend in the difference of serum total ΔPCS was independent of eGFR and dietary protein:fiber ratio intake. No difference was found in urinary PCS. Aside from the decreased high-density lipoprotein cholesterol in the intervention, no differences were observed in the change of IS, IAA or other secondary outcome between the groups. CONCLUSIONS: Our result suggests the potential of FOS in reducing serum total and free PCS in nondiabetic NDD-CKD patients.


Subject(s)
Gastrointestinal Tract/drug effects , Microbiota/physiology , Oligosaccharides/administration & dosage , Prebiotics/administration & dosage , Renal Insufficiency, Chronic/drug therapy , Toxins, Biological/isolation & purification , Uremia/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cresols/blood , Dietary Proteins , Double-Blind Method , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Glomerular Filtration Rate , Humans , Inflammation/prevention & control , Male , Microbiota/drug effects , Middle Aged , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/microbiology , Toxins, Biological/metabolism , Uremia/microbiology , Young Adult
20.
J Nephrol ; 32(1): 27-37, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30069677

ABSTRACT

In chronic kidney disease (CKD), the progressive decline in the renal excretory function leads to accumulation of urea and toxins in the blood. The CKD-associated dysbiosis of gut microbiota further contributes to uremia by increasing intestinal toxins production. Gut microbiota is involved in a complex network of human organs, mediated by microbial metabolites: in CKD, gut-heart and gut-brain axes may have a role in increased cardiovascular risk and neuropsychiatric disorders. While the cardiovascular toxicity of some microbial molecules is well known, their presumptive neurotoxicity needs to be confirmed by specific studies. In this review, we describe gut-heart and gut-brain axes in CKD, with an overview of the experimental and human studies characterizing CKD-associated gut microbiota, and we discuss the benefits coming from new approaches aimed at gut manipulation. Microbiota metabolism is emerging as a modifiable non-traditional risk factor in nephrology. In order to take advantage of this issue, it is necessary to consider the microbiota manipulation as part of the nutritional management of CKD. Integrating the low-protein nutritional approach with prebiotic, probiotic and synbiotic supplementation is a promising tool to control disease progression and comorbidities, though an extensive validation in large-scale clinical trials is still required.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Kidney/physiopathology , Renal Elimination , Renal Insufficiency, Chronic/microbiology , Urea/blood , Uremia/microbiology , Animals , Brain/metabolism , Brain/physiopathology , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Diet, Protein-Restricted , Dysbiosis , Host-Pathogen Interactions , Humans , Prebiotics , Probiotics/therapeutic use , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diet therapy , Renal Insufficiency, Chronic/physiopathology , Synbiotics , Uremia/blood , Uremia/diet therapy , Uremia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...